1
|
Bean L, Bose PK, Rani A, Kumar A. Serine racemase expression profile in the prefrontal cortex and hippocampal subregions during aging in male and female rats. Aging (Albany NY) 2024; 16:8402-8416. [PMID: 38761177 PMCID: PMC11164512 DOI: 10.18632/aging.205841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/10/2024] [Indexed: 05/20/2024]
Abstract
Aging is associated with a decrease in N-methyl-D-aspartate (NMDA) receptor function, which is critical for maintaining synaptic plasticity, learning, and memory. Activation of the NMDA receptor requires binding of the neurotransmitter glutamate and also the presence of co-agonist D-serine at the glycine site. The enzymatic conversion of L-serine to D-serine is facilitated by the enzyme serine racemase (SR). Subsequently, SR plays a pivotal role in regulating NMDA receptor activity, thereby impacting synaptic plasticity and memory processes in the central nervous system. As such, age-related changes in the expression of SR could contribute to decreased NMDA receptor function. However, age-associated changes in SR expression levels in the medial and lateral prefrontal cortex (mPFC, lPFC), and in the dorsal hippocampal subfields, CA1, CA3, and dentate gyrus (DG), have not been thoroughly elucidated. Therefore, the current studies were designed to determine the SR expression profile, including protein levels and mRNA, for these regions in aged and young male and female Fischer-344 rats. Our results demonstrate a significant reduction in SR expression levels in the mPFC and all hippocampal subfields of aged rats compared to young rats. No sex differences were observed in the expression of SR. These findings suggest that the decrease in SR levels may play a role in the age-associated reduction of NMDA receptor function in brain regions crucial for cognitive function and synaptic plasticity.
Collapse
Affiliation(s)
- Linda Bean
- Department of Anatomy, Cell Biology, and Physiology, IU School of Medicine, Indianapolis, IN 46201, USA
| | - Prodip K. Bose
- Brain Rehabilitation Research Center, Malcom Randall Department of Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32607, USA
- Department of Anesthesiology and Department of Neurology at the College of Medicine, University of Florida, FL 32607, USA
| | - Asha Rani
- Department of Neuroscience, The McKnight Brain Institute, University of Florida, Gainesville, FL 32607, USA
| | - Ashok Kumar
- Department of Neuroscience, The McKnight Brain Institute, University of Florida, Gainesville, FL 32607, USA
| |
Collapse
|
2
|
Zarrella JA, Tsurumi A. Genome-wide transcriptome profiling and development of age prediction models in the human brain. Aging (Albany NY) 2024; 16:4075-4094. [PMID: 38428408 PMCID: PMC10968712 DOI: 10.18632/aging.205609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 03/28/2023] [Indexed: 03/03/2024]
Abstract
Aging-related transcriptome changes in various regions of the healthy human brain have been explored in previous works, however, a study to develop prediction models for age based on the expression levels of specific panels of transcripts is lacking. Moreover, studies that have assessed sexually dimorphic gene activities in the aging brain have reported discrepant results, suggesting that additional studies would be advantageous. The prefrontal cortex (PFC) region was previously shown to have a particularly large number of significant transcriptome alterations during healthy aging in a study that compared different regions in the human brain. We harmonized neuropathologically normal PFC transcriptome datasets obtained from the Gene Expression Omnibus (GEO) repository, ranging in age from 21 to 105 years, and found a large number of differentially regulated transcripts in the old and elderly, compared to young samples overall, and compared female and male-specific expression alterations. We assessed the genes that were associated with age by employing ontology, pathway, and network analyses. Furthermore, we applied various established (least absolute shrinkage and selection operator (Lasso) and Elastic Net (EN)) and recent (eXtreme Gradient Boosting (XGBoost) and Light Gradient Boosting Machine (LightGBM)) machine learning algorithms to develop accurate prediction models for chronological age and validated them. Studies to further validate these models in other large populations and molecular studies to elucidate the potential mechanisms by which the transcripts identified may be related to aging phenotypes would be advantageous.
Collapse
Affiliation(s)
- Joseph A. Zarrella
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Shriner's Hospitals for Children-Boston, Boston, MA 02114, USA
| |
Collapse
|
3
|
Budamagunta V, Kumar A, Rani A, Manohar Sindhu S, Yang Y, Zhou D, Foster TC. Senolytic treatment alleviates doxorubicin-induced chemobrain. Aging Cell 2024; 23:e14037. [PMID: 38225896 PMCID: PMC10861213 DOI: 10.1111/acel.14037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 01/17/2024] Open
Abstract
Doxorubicin (Dox), a widely used treatment for cancer, can result in chemotherapy-induced cognitive impairments (chemobrain). Chemobrain is associated with inflammation and oxidative stress similar to aging. As such, Dox treatment has also been used as a model of aging. However, it is unclear if Dox induces brain changes similar to that observed during aging since Dox does not readily enter the brain. Rather, the mechanism for chemobrain likely involves the induction of peripheral cellular senescence and the release of senescence-associated secretory phenotype (SASP) factors and these SASP factors can enter the brain to disrupt cognition. We examined the effect of Dox on peripheral and brain markers of aging and cognition. In addition, we employed the senolytic, ABT-263, which also has limited access to the brain. The results indicate that plasma SASP factors enter the brain, activating microglia, increasing oxidative stress, and altering gene transcription. In turn, the synaptic function required for memory was reduced in response to altered redox signaling. ABT-263 prevented or limited most of the Dox-induced effects. The results emphasize a link between cognitive decline and the release of SASP factors from peripheral senescent cells and indicate some differences as well as similarities between advanced age and Dox treatment.
Collapse
Affiliation(s)
- Vivekananda Budamagunta
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Sahana Manohar Sindhu
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Yang Yang
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Daohong Zhou
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Department of Biochemistry and Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
4
|
Gandy HM, Hollis F, Hernandez CM, McQuail JA. Aging or chronic stress impairs working memory and modulates GABA and glutamate gene expression in prelimbic cortex. Front Aging Neurosci 2024; 15:1306496. [PMID: 38259638 PMCID: PMC10800675 DOI: 10.3389/fnagi.2023.1306496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
The glucocorticoid (GC) hypothesis posits that effects of stress and dysregulated hypothalamic-pituitary-adrenal axis activity accumulate over the lifespan and contribute to impairment of neural function and cognition in advanced aging. The validity of the GC hypothesis is bolstered by a wealth of studies that investigate aging of the hippocampus and decline of associated mnemonic functions. The prefrontal cortex (PFC) mediates working memory which also decreases with age. While the PFC is susceptible to stress and GCs, few studies have formally assessed the application of the GC hypothesis to PFC aging and working memory. Using parallel behavioral and molecular approaches, we compared the effects of normal aging versus chronic variable stress (CVS) on working memory and expression of genes that encode for effectors of glutamate and GABA signaling in male F344 rats. Using an operant delayed match-to-sample test of PFC-dependent working memory, we determined that normal aging and CVS each significantly impaired mnemonic accuracy and reduced the total number of completed trials. We then determined that normal aging increased expression of Slc6a11, which encodes for GAT-3 GABA transporter expressed by astrocytes, in the prelimbic (PrL) subregion of the PFC. CVS increased PrL expression of genes associated with glutamatergic synapses: Grin2b that encodes the GluN2B subunit of NMDA receptor, Grm4 that encodes for metabotropic glutamate receptor 4 (mGluR4), and Plcb1 that encodes for phospholipase C beta 1, an intracellular signaling enzyme that transduces signaling of Group I mGluRs. Beyond the identification of specific genes that were differentially expressed between the PrL in normal aging or CVS, examination of Log2 fold-changes for all expressed glutamate and GABA genes revealed a positive association between molecular phenotypes of aging and CVS in the PrL but no association in the infralimbic subregion. Consistent with predictions of the GC hypothesis, PFC-dependent working memory and PrL glutamate/GABA gene expression demonstrate comparable sensitivity to aging and chronic stress. However, changes in expression of specific genes affiliated with regulation of extracellular GABA in normal aging vs. genes encoding for effectors of glutamatergic signaling during CVS suggest the presence of unique manifestations of imbalanced inhibitory and excitatory signaling in the PFC.
Collapse
Affiliation(s)
- Hannah M. Gandy
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Health Care System, Columbia, SC, United States
| | - Caesar M. Hernandez
- Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, The University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Joseph A. McQuail
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Cox LA, Puppala S, Chan J, Zimmerman KD, Hamid Z, Ampong I, Huber HF, Li G, Jadhav AYL, Wang B, Li C, Baxter MG, Shively C, Clarke GD, Register TC, Nathanielsz PW, Olivier M. Integrated multi-omics analysis of brain aging in female nonhuman primates reveals altered signaling pathways relevant to age-related disorders. Neurobiol Aging 2023; 132:109-119. [PMID: 37797463 PMCID: PMC10841409 DOI: 10.1016/j.neurobiolaging.2023.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 10/07/2023]
Abstract
The prefrontal cortex (PFC) has been implicated as a key brain region responsible for age-related cognitive decline. Little is known about aging-related molecular changes in PFC that may mediate these effects. To date, no studies have used untargeted discovery methods with integrated analyses to determine PFC molecular changes in healthy female primates. We quantified PFC changes associated with healthy aging in female baboons by integrating multiple omics data types (transcriptomics, proteomics, metabolomics) from samples across the adult age span. Our integrated omics approach using unbiased weighted gene co-expression network analysis to integrate data and treat age as a continuous variable, revealed highly interconnected known and novel pathways associated with PFC aging. We found Gamma-aminobutyric acid (GABA) tissue content associated with these signaling pathways, providing 1 potential biomarker to assess PFC changes with age. These highly coordinated pathway changes during aging may represent early steps for aging-related decline in PFC functions, such as learning and memory, and provide potential biomarkers to assess cognitive status in humans.
Collapse
Affiliation(s)
- Laura A Cox
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA.
| | - Sobha Puppala
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jeannie Chan
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Zeeshan Hamid
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Isaac Ampong
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Hillary F Huber
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ge Li
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Avinash Y L Jadhav
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Benlian Wang
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Cun Li
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Mark G Baxter
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Carol Shively
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Geoffrey D Clarke
- Department of Radiology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Thomas C Register
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Peter W Nathanielsz
- Texas Pregnancy & Life-Course Health Research Center, Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Michael Olivier
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
6
|
Zheng QM, Zhou ZR, Hou XY, Lv N, Zhang YQ, Cao H. Transcriptome Analysis of the Mouse Medial Prefrontal Cortex in a Chronic Constriction Injury Model. Neuromolecular Med 2023; 25:375-387. [PMID: 36971954 DOI: 10.1007/s12017-023-08742-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023]
Abstract
The medial prefrontal cortex (mPFC) is critical for both the sensory and emotional/cognitive components of pain. However, the underlying mechanism remains largely unknown. Here, we examined changes in the transcriptomic profiles in the mPFC of mice with chronic pain using RNA sequencing (RNA-seq) technology. A mouse model of peripheral neuropathic pain was established via chronic constriction injury (CCI) of the sciatic nerve. CCI mice developed sustained mechanical allodynia and thermal hyperalgesia, as well as cognitive impairment four weeks after surgery. RNA-seq was conducted 4 weeks after CCI surgery. Compared with contral group, RNA-seq identified a total 309 and 222 differentially expressed genes (DEGs) in the ipsilateral and contralateral mPFC of CCI model mice, respectively. GO analysis indicated that the functions of these genes were mainly enriched in immune- and inflammation-related processes such as interferon-gamma production and cytokine secretion. KEGG analysis further showed the enrichment of genes involved in the neuroactive ligand-receptor interaction signaling pathway and Parkinson disease pathway that have been reported to be importantly involved in chronic neuralgia and cognitive dysfunction. Our study may provide insights into the possible mechanisms underlying neuropathic pain and pain-related comorbidities.
Collapse
Affiliation(s)
- Qi-Min Zheng
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zi-Rui Zhou
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xin-Yu Hou
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ning Lv
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hong Cao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Li Y, Khan N, Ritzel RM, Lei Z, Allen S, Faden AI, Wu J. Sexually dimorphic extracellular vesicle responses after chronic spinal cord injury are associated with neuroinflammation and neurodegeneration in the aged brain. J Neuroinflammation 2023; 20:197. [PMID: 37653491 PMCID: PMC10469550 DOI: 10.1186/s12974-023-02881-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Medical advances have made it increasingly possible for spinal cord injury (SCI) survivors to survive decades after the insult. But how SCI affects aging changes and aging impacts the injury process have received limited attention. Extracellular vesicles (EVs) are recognized as critical mediators of neuroinflammation after CNS injury, including at a distance from the lesion site. We have previously shown that SCI in young male mice leads to robust changes in plasma EV count and microRNA (miR) content. Here, our goal was to investigate the impact of biological sex and aging on EVs and brain after SCI. METHODS Young adult age-matched male and female C57BL/6 mice were subjected to SCI. At 19 months post-injury, total plasma EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis (NTA). EVs miR cargo was examined using the Fireplex® assay. The transcriptional changes in the brain were assessed by a NanoString nCounter Neuropathology panel and validated by Western blot (WB) and flow cytometry (FC). A battery of behavioral tests was performed for assessment of neurological function. RESULTS Transcriptomic changes showed a high number of changes between sham and those with SCI. Sex-specific changes were found in transcription networks related to disease association, activated microglia, and vesicle trafficking. FC showed higher microglia and myeloid counts in the injured tissue of SCI/Female compared to their male counterparts, along with higher microglial production of ROS in both injured site and the brain. In the latter, increased levels of TNF and mitochondrial membrane potential were seen in microglia from SCI/Female. WB and NTA revealed that EV markers are elevated in the plasma of SCI/Male. Particle concentration in the cortex increased after injury, with SCI/Female showing higher counts than SCI/Male. EVs cargo analysis revealed changes in miR content related to injury and sex. Behavioral testing confirmed impairment of cognition and depression at chronic time points after SCI in both sexes, without significant differences between males and females. CONCLUSIONS Our study is the first to show sexually dimorphic changes in brain after very long-term SCI and supports a potential sex-dependent EV-mediated mechanism that contributes to SCI-induced brain changes.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Niaz Khan
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Samantha Allen
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
Budamagunta V, Kumar A, Rani A, Bean L, Manohar‐Sindhu S, Yang Y, Zhou D, Foster TC. Effect of peripheral cellular senescence on brain aging and cognitive decline. Aging Cell 2023; 22:e13817. [PMID: 36959691 PMCID: PMC10186609 DOI: 10.1111/acel.13817] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023] Open
Abstract
We examine similar and differential effects of two senolytic treatments, ABT-263 and dasatinib + quercetin (D + Q), in preserving cognition, markers of peripheral senescence, and markers of brain aging thought to underlie cognitive decline. Male F344 rats were treated from 12 to 18 months of age with D + Q, ABT-263, or vehicle, and were compared to young (6 months). Both senolytic treatments rescued memory, preserved the blood-brain barrier (BBB) integrity, and prevented the age-related decline in hippocampal N-methyl-D-aspartate receptor (NMDAR) function associated with impaired cognition. Senolytic treatments decreased senescence-associated secretory phenotype (SASP) and inflammatory cytokines/chemokines in the plasma (IL-1β, IP-10, and RANTES), with some markers more responsive to D + Q (TNFα) or ABT-263 (IFNγ, leptin, EGF). ABT-263 was more effective in decreasing senescence genes in the spleen. Both senolytic treatments decreased the expression of immune response and oxidative stress genes and increased the expression of synaptic genes in the dentate gyrus (DG). However, D + Q influenced twice as many genes as ABT-263. Relative to D + Q, the ABT-263 group exhibited increased expression of DG genes linked to cell death and negative regulation of apoptosis and microglial cell activation. Furthermore, D + Q was more effective at decreasing morphological markers of microglial activation. The results indicate that preserved cognition was associated with the removal of peripheral senescent cells, decreasing systemic inflammation that normally drives neuroinflammation, BBB breakdown, and impaired synaptic function. Dissimilarities associated with brain transcription indicate divergence in central mechanisms, possibly due to differential access.
Collapse
Affiliation(s)
- Vivekananda Budamagunta
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Linda Bean
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Sahana Manohar‐Sindhu
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Yang Yang
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Department of Biochemistry and Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Daohong Zhou
- Department of Biochemistry and Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
9
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
10
|
Myrum C, Moreno-Castilla P, Rapp PR. 'Arc'-hitecture of normal cognitive aging. Ageing Res Rev 2022; 80:101678. [PMID: 35781092 PMCID: PMC9378697 DOI: 10.1016/j.arr.2022.101678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 12/17/2022]
Abstract
Arc is an effector immediate-early gene that is critical for forming long-term memories. Since its discovery 25 years ago, it has repeatedly surprised us with a number of intriguing properties, including the transport of its mRNA to recently-activated synapses, its master role in bidirectionally regulating synaptic strength, its evolutionary retroviral origins, its ability to mediate intercellular transfer between neurons via extracellular vesicles (EVs), and its exceptional regulation-both temporally and spatially. The current review discusses how Arc has been used as a tool to identify the neural networks involved in cognitive aging and how Arc itself may contribute to cognitive outcome in aging. In addition, we raise several outstanding questions, including whether Arc-containing EVs in peripheral blood might provide a noninvasive biomarker for memory-related synaptic failure in aging, and whether rectifying Arc dysregulation is likely to be an effective strategy for bending the arc of aging toward successful cognitive outcomes.
Collapse
Affiliation(s)
- Craig Myrum
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Perla Moreno-Castilla
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Peter R Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
11
|
Dietary Restriction and Rapamycin Affect Brain Aging in Mice by Attenuating Age-Related DNA Methylation Changes. Genes (Basel) 2022; 13:genes13040699. [PMID: 35456505 PMCID: PMC9030181 DOI: 10.3390/genes13040699] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023] Open
Abstract
The fact that dietary restriction (DR) and long-term rapamycin treatment (RALL) can ameliorate the aging process has been reported by many researchers. As the interface between external and genetic factors, epigenetic modification such as DNA methylation may have latent effects on the aging rate at the molecular level. To understand the mechanism behind the impacts of dietary restriction and rapamycin on aging, DNA methylation and gene expression changes were measured in the hippocampi of different-aged mice. Examining the single-base resolution of DNA methylation, we discovered that both dietary restriction and rapamycin treatment can maintain DNA methylation in a younger state compared to normal-aged mice. Through functional enrichment analysis of genes in which DNA methylation or gene expression can be affected by DR/RALL, we found that DR/RALL may retard aging through a relationship in which DNA methylation and gene expression work together not only in the same gene but also in the same biological process. This study is instructive for understanding the maintenance of DNA methylation by DR/RALL in the aging process, as well as the role of DR and RALL in the amelioration of aging.
Collapse
|
12
|
Zheng Y, Habes M, Gonzales M, Pomponio R, Nasrallah I, Khan S, Vaughan DE, Davatzikos C, Seshadri S, Launer L, Sorond F, Sedaghat S, Wainwright D, Baccarelli A, Sidney S, Bryan N, Greenland P, Lloyd-Jones D, Yaffe K, Hou L. Mid-life epigenetic age, neuroimaging brain age, and cognitive function: coronary artery risk development in young adults (CARDIA) study. Aging (Albany NY) 2022; 14:1691-1712. [PMID: 35220276 PMCID: PMC8908939 DOI: 10.18632/aging.203918] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022]
Abstract
The proportion of aging populations affected by dementia is increasing. There is an urgent need to identify biological aging markers in mid-life before symptoms of age-related dementia present for early intervention to delay the cognitive decline and the onset of dementia. In this cohort study involving 1,676 healthy participants (mean age 40) with up to 15 years of follow up, we evaluated the associations between cognitive function and two classes of novel biological aging markers: blood-based epigenetic aging and neuroimaging-based brain aging. Both accelerated epigenetic aging and brain aging were prospectively associated with worse cognitive outcomes. Specifically, every year faster epigenetic or brain aging was on average associated with 0.19-0.28 higher (worse) Stroop score, 0.04-0.05 lower (worse) RAVLT score, and 0.23-0.45 lower (worse) DSST (all false-discovery-rate-adjusted p <0.05). While epigenetic aging is a more stable biomarker with strong long-term predictive performance for cognitive function, brain aging biomarker may change more dynamically in temporal association with cognitive decline. The combined model using epigenetic and brain aging markers achieved the highest accuracy (AUC: 0.68, p<0.001) in predicting global cognitive function status. Accelerated epigenetic age and brain age at midlife may aid timely identification of individuals at risk for accelerated cognitive decline and promote the development of interventions to preserve optimal functioning across the lifespan.
Collapse
Affiliation(s)
- Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mohamad Habes
- Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitzi Gonzales
- Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Raymond Pomponio
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ilya Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sadiya Khan
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Douglas E. Vaughan
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Christos Davatzikos
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sudha Seshadri
- Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lenore Launer
- Laboratory of Epidemiology and Population Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Farzaneh Sorond
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Derek Wainwright
- Departments of Neurological Surgery, Medicine-Hematology and Oncology, Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Stephen Sidney
- Kaiser Permanente Division of Research, Oakland, CA 94612, USA
| | - Nick Bryan
- Department of Diagnostic Medicine, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Philip Greenland
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Donald Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kristine Yaffe
- Departments of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
- Department of Neurology University of California, San Francisco, CA 94143, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, CA 94143, USA
- San Francisco VA Medical Center, San Francisco, CA 94143, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
13
|
Rani A, Barter J, Kumar A, Stortz JA, Hollen M, Nacionales D, Moldawer LL, Efron PA, Foster TC. Influence of age and sex on microRNA response and recovery in the hippocampus following sepsis. Aging (Albany NY) 2022; 14:728-746. [PMID: 35094981 PMCID: PMC8833110 DOI: 10.18632/aging.203868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022]
Abstract
Sepsis, defined as a dysregulated host immune response to infection, is a common and dangerous clinical syndrome. The excessive host inflammatory response can induce immediate and persistent cognitive decline, which can be worse in older individuals. Sex-specific differences in the outcome of infectious diseases and sepsis appear to favor females. We employed a murine model to examine the influence of age and sex on the brain's microRNA (miR) response following sepsis. Young and old mice of both sexes underwent cecal ligation and puncture (CLP) with daily restraint stress. Expression of hippocampal miR was examined in age- and sex-matched controls at 1 and 4 days post-CLP. Few miR were modified in a similar manner across age or sex and these few miR were generally associated with neuroprotection against inflammation. Similar to previous work examining transcription, young females exhibited a better recovery of the miR profile from day 1 to day 4, relative to young males and old females. For young males and all female groups, the initial response mainly involved a decrease in miR expression. In contrast, old males exhibited only upregulated miR on day 1 and day 4 and many of the miR upregulated on day 1 and day 4 were linked to neurodegeneration, increased neuroinflammation, and cognitive impairment. The results emphasize age and sex differences in epigenetic mechanisms that likely contribute to susceptibility or resilience to cognitive impairment due to sepsis.
Collapse
Affiliation(s)
- Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jolie Barter
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Julie A Stortz
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - McKenzie Hollen
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Dina Nacionales
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Philip A Efron
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.,Genetics and Genomics Program, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
14
|
Adulthood systemic inflammation accelerates the trajectory of age-related cognitive decline. Aging (Albany NY) 2021; 13:22092-22108. [PMID: 34587117 PMCID: PMC8507275 DOI: 10.18632/aging.203588] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022]
Abstract
In order to understand the long-term effects of systemic inflammation, it is important to distinguish inflammation-induced changes in baseline cognitive function from changes that interact with aging to influence the trajectory of cognitive decline. Lipopolysaccharide (LPS; 1 mg/kg) or vehicle was administered to young adult (6 months) male rats via intraperitoneal injections, once a week for 7 weeks. Longitudinal effects on cognitive decline were examined 6 and 12 months after the initial injections. Repeated LPS treatment, in adults, resulted in a long-term impairment in memory, examined in aged animals (age 18 months), but not in middle-age (age 12 months). At 12 months following injections, LPS treatment was associated with a decrease in N-methyl-D-aspartate receptor-mediated component of synaptic transmission and altered expression of genes linked to the synapse and to regulation of the response to inflammatory signals. The results of the current study suggest that the history of systemic inflammation is one component of environmental factors that contribute to the resilience or susceptibility to age-related brain changes and associated trajectory of cognitive decline.
Collapse
|
15
|
Yegla B, Foster TC. Operationally defining cognitive reserve genes. Neurobiol Aging 2021; 110:96-105. [PMID: 34565615 DOI: 10.1016/j.neurobiolaging.2021.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/05/2021] [Accepted: 08/24/2021] [Indexed: 01/22/2023]
Abstract
Variability in cognitive decline is related to the environment, lifestyle factors, and individual differences in biological aging, including cognitive reserve, plastic properties of the brain, which account for better-than-expected cognition for a given level of brain aging or pathology. Cognitive reserve has not been thoroughly investigated in aged rodents. To address this gap, cognitive reserve was examined using Gene Expression Omnibus data for the CA1 region of the hippocampus of young and aged behaviorally characterized male rats. Statistical filtering identified brain aging and potential cognitive reserve genes, and multiple regression was employed to confirm cognitive reserve genes as genes that predicted better-than-expected cognition for a given level of brain aging. In general, cognitive reserve genes, in which increased expression was associated with better cognition, were not different with age or directly correlated with measures of cognition and appear to act as negative regulators of aging processes, including neuroinflammation and oxidative stress. The results suggest that, for some animals, resilience mechanisms are activated to counteract aging stressors that impair cognition. In contrast, cognitive reserve genes, in which decreased expression was associated with better cognition, were linked to nervous system development and cation transport, suggesting adaptive changes in the circuit to preserve cognition.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Genetics and Genomics Program University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Gulmez Karaca K, Brito DVC, Kupke J, Zeuch B, Oliveira AMM. Engram reactivation during memory retrieval predicts long-term memory performance in aged mice. Neurobiol Aging 2021; 101:256-261. [PMID: 33647524 DOI: 10.1016/j.neurobiolaging.2021.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/20/2021] [Indexed: 10/22/2022]
Abstract
Age-related cognitive decline preferentially targets long-lasting episodic memories that require intact hippocampal function. Memory traces (or engrams) are believed to be encoded within the neurons activated during learning (neuronal ensembles), and recalled by reactivation of the same population. However, whether engram reactivation dictates memory performance late in life is not known. Here, we labeled neuronal ensembles formed during object location recognition learning in the dentate gyrus, and analyzed the reactivation of this population during long-term memory recall in young adult, cognitively impaired- and unimpaired-aged mice. We found that reactivation of memory-encoding neuronal ensembles at long-term memory recall was disrupted in impaired but not unimpaired-aged mice. Furthermore, we showed that the memory performance in the aged population correlated with the degree of engram reactivation at long-term memory recall. Overall, our data implicates recall-induced engram reactivation as a prediction factor of memory performance in aging. Moreover, our findings suggest impairments in neuronal ensemble stabilization and/or reactivation as an underlying mechanism in age-dependent cognitive decline.
Collapse
Affiliation(s)
- Kubra Gulmez Karaca
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - David V C Brito
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Benjamin Zeuch
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
17
|
McQuail JA, Dunn AR, Stern Y, Barnes CA, Kempermann G, Rapp PR, Kaczorowski CC, Foster TC. Cognitive Reserve in Model Systems for Mechanistic Discovery: The Importance of Longitudinal Studies. Front Aging Neurosci 2021; 12:607685. [PMID: 33551788 PMCID: PMC7859530 DOI: 10.3389/fnagi.2020.607685] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
The goal of this review article is to provide a resource for longitudinal studies, using animal models, directed at understanding and modifying the relationship between cognition and brain structure and function throughout life. We propose that forthcoming longitudinal studies will build upon a wealth of knowledge gleaned from prior cross-sectional designs to identify early predictors of variability in cognitive function during aging, and characterize fundamental neurobiological mechanisms that underlie the vulnerability to, and the trajectory of, cognitive decline. Finally, we present examples of biological measures that may differentiate mechanisms of the cognitive reserve at the molecular, cellular, and network level.
Collapse
Affiliation(s)
- Joseph A. McQuail
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Amy R. Dunn
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Yaakov Stern
- Cognitive Neuroscience Division, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Carol A. Barnes
- Departments of Psychology and Neuroscience, University of Arizona, Tucson, AZ, United States
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, United States
| | - Gerd Kempermann
- CRTD—Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers (HZ), Dresden, Germany
| | - Peter R. Rapp
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging, Baltimore, MD, United States
| | | | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
18
|
Linial M, Stern A, Weinstock M. Effect of ladostigil treatment of aging rats on gene expression in four brain areas associated with regulation of memory. Neuropharmacology 2020; 177:108229. [PMID: 32738309 DOI: 10.1016/j.neuropharm.2020.108229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/17/2020] [Accepted: 07/01/2020] [Indexed: 02/09/2023]
Abstract
Episodic and spatial memory decline in aging and are controlled by the hippocampus, perirhinal, frontal and parietal cortices and the connections between them. Ladostigil, a drug with antioxidant and anti-inflammatory activity, was shown to prevent the loss of episodic and spatial memory in aging rats. To better understand the molecular effects of aging and ladostigil on these brain regions we characterized the changes in gene expression using RNA-sequencing technology in rats aged 6 and 22 months. We found that the changes induced by aging and chronic ladostigil treatment were brain region specific. In the hippocampus, frontal and perirhinal cortex, ladostigil decreased the overexpression of genes regulating calcium homeostasis, ion channels and those adversely affecting synaptic function. In the parietal cortex, ladostigil increased the expression of several genes that provide neurotrophic support, while reducing that of pro-apoptotic genes and those encoding pro-inflammatory cytokines and their receptors. Ladostigil also decreased the expression of axonal growth inhibitors and those impairing mitochondrial function. Together, these actions could explain the protection by ladostigil against age-related memory decline.
Collapse
Affiliation(s)
- Michal Linial
- Department of Biological Chemistry, Life Science Institute, Israel; The Rachel and Selim Benin School of Computer Science and Engineering, Israel
| | - Amos Stern
- Department of Biological Chemistry, Life Science Institute, Israel
| | - Marta Weinstock
- Institute of Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
19
|
Hahn A, Pensold D, Bayer C, Tittelmeier J, González-Bermúdez L, Marx-Blümel L, Linde J, Groß J, Salinas-Riester G, Lingner T, von Maltzahn J, Spehr M, Pieler T, Urbach A, Zimmer-Bensch G. DNA Methyltransferase 1 (DNMT1) Function Is Implicated in the Age-Related Loss of Cortical Interneurons. Front Cell Dev Biol 2020; 8:639. [PMID: 32793592 PMCID: PMC7387673 DOI: 10.3389/fcell.2020.00639] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/25/2020] [Indexed: 01/19/2023] Open
Abstract
Increased life expectancy in modern society comes at the cost of age-associated disabilities and diseases. Aged brains not only show reduced excitability and plasticity, but also a decline in inhibition. Age-associated defects in inhibitory circuits likely contribute to cognitive decline and age-related disorders. Molecular mechanisms that exert epigenetic control of gene expression contribute to age-associated neuronal impairments. Both DNA methylation, mediated by DNA methyltransferases (DNMTs), and histone modifications maintain neuronal function throughout lifespan. Here we provide evidence that DNMT1 function is implicated in the age-related loss of cortical inhibitory interneurons. Dnmt1 deletion in parvalbumin-positive interneurons attenuates their age-related decline in the cerebral cortex. Moreover, conditional Dnmt1-deficient mice show improved somatomotor performance and reduced aging-associated transcriptional changes. A decline in the proteostasis network, responsible for the proper degradation and removal of defective proteins, is implicated in age- and disease-related neurodegeneration. Our data suggest that DNMT1 acts indirectly on interneuron survival in aged mice by modulating the proteostasis network during life-time.
Collapse
Affiliation(s)
- Anne Hahn
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Daniel Pensold
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany.,Department of Functional Epigenetics in the Animal Model, Institute of Biology II, RWTH Aachen University, Aachen, Germany
| | - Cathrin Bayer
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany.,Department of Functional Epigenetics in the Animal Model, Institute of Biology II, RWTH Aachen University, Aachen, Germany
| | - Jessica Tittelmeier
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Lourdes González-Bermúdez
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Lisa Marx-Blümel
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Jenice Linde
- Department of Functional Epigenetics in the Animal Model, Institute of Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| | - Jonas Groß
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany
| | - Gabriela Salinas-Riester
- Transcriptome and Genome Analysis Laboratory (TAL), Department of Developmental Biochemistry, University of Göttingen, Göttingen, Germany
| | - Thomas Lingner
- Transcriptome and Genome Analysis Laboratory (TAL), Department of Developmental Biochemistry, University of Göttingen, Göttingen, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Marc Spehr
- Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany.,Department of Chemosensation, Institute of Biology II, RWTH Aachen University, Aachen, Germany
| | - Tomas Pieler
- Centre for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Department of Developmental Biochemistry, University of Göttingen, Göttingen, Germany
| | - Anja Urbach
- Institute of Neurology, University Hospital Jena, Jena, Germany
| | - Geraldine Zimmer-Bensch
- Department of Functional Epigenetics, Institute of Human Genetics, University Hospital Jena, Jena, Germany.,Department of Functional Epigenetics in the Animal Model, Institute of Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
20
|
Barter J, Kumar A, Rani A, Colon-Perez LM, Febo M, Foster TC. Differential Effect of Repeated Lipopolysaccharide Treatment and Aging on Hippocampal Function and Biomarkers of Hippocampal Senescence. Mol Neurobiol 2020; 57:4045-4059. [PMID: 32651758 DOI: 10.1007/s12035-020-02008-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023]
Abstract
Markers of brain aging and cognitive decline are thought to be influenced by peripheral inflammation. This study compared the effects of repeated lipopolysaccharide (LPS) treatment in young rats to age-related changes in hippocampal-dependent cognition and transcription. Young Fischer 344 X Brown Norway hybrid rats were given intraperitoneal injections once a week for 7 weeks with either LPS or vehicle. Older rats received a similar injection schedule of vehicle. Old vehicle and young LPS rats exhibited a delay-dependent impairment in spatial memory. Further, LPS treatment reduced the hippocampal CA3-CA1 synaptic response. RNA sequencing, performed on CA1, indicated an increase in genes linked to neuroinflammation in old vehicle and young LPS animals. In contrast to an age-related decrease in transcription of synaptic genes, young LPS animals exhibited increased expression of genes that support the growth and maintenance of synapses. We suggest that the increased expression of genes for growth and maintenance of synapses in young animals represents neuronal resilience/recovery in response to acute systemic inflammation. Thus, the results indicate that repeated LPS treatment does not completely recapitulate the aging phenotype for synaptic function, possibly due to the chronic nature of systemic inflammation in aging and resilience of young animals to acute treatments.
Collapse
Affiliation(s)
- Jolie Barter
- Department of Medicine, Division of General Medicine and Geriatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610-0244, USA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610-0244, USA
| | - Luis M Colon-Perez
- Department of Neurobiology and Behavior, Center for Learning and Memory, University of California, Irvine, CA, 92697, USA
| | - Marcelo Febo
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610-0244, USA.,Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,Center for Addiction Research and Education, University of Florida, Gainesville, FL, 32611, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32610-0244, USA. .,Genetics and Genomics Program, University of Florida, Gainesville, 32611, FL, USA.
| |
Collapse
|
21
|
Hippocampal Subregion Transcriptomic Profiles Reflect Strategy Selection during Cognitive Aging. J Neurosci 2020; 40:4888-4899. [PMID: 32376783 DOI: 10.1523/jneurosci.2944-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/08/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Age-related cognitive impairments are associated with differentially expressed genes (DEGs) linked to defined neural systems; however, studies examining multiple regions of the hippocampus fail to find links between behavior and transcription in the dentate gyrus (DG). We hypothesized that use of a task requiring intact DG function would emphasize molecular signals in the DG associated with a decline in performance. We used a water maze beacon discrimination task to characterize young and middle-age male F344 rats, followed by a spatial reference memory probe trial test. Middle-age rats showed increased variability in discriminating two identical beacons. Use of an allocentric strategy and formation of a spatial reference memory were not different between age groups; however, older animals compensated for impaired beacon discrimination through greater reliance on spatial reference memory. mRNA sequencing of hippocampal subregions indicated DEGs in the DG of middle-age rats, linked to synaptic function and neurogenesis, correlated with beacon discrimination performance, suggesting that senescence of the DG underlies the impairment. Few genes correlated with spatial memory across age groups, with a greater number in region CA1. Age-related CA1 DEGs, correlated with spatial memory, were linked to regulation of neural activity. These results indicate that the beacon task is sensitive to impairment in middle age, and distinct gene profiles are observed in neural circuits that underlie beacon discrimination performance and allocentric memory. The use of different strategies in older animals and associated transcriptional profiles could provide an animal model for examining cognitive reserve and neural compensation of aging.SIGNIFICANCE STATEMENT Hippocampal subregions are thought to differentially contribute to memory. We took advantage of age-related variability in performance on a water maze beacon task and next-generation sequencing to test the hypothesis that aging of the dentate gyrus is linked to impaired beacon discrimination and compensatory use of allocentric memory. The dentate gyrus expressed synaptic function and neurogenesis genes correlated with beacon discrimination in middle-age animals. Spatial reference memory was associated with CA1 transcriptional correlates linked to regulation of neural activity and use of an allocentric strategy. This is the first study examining transcriptomes of multiple hippocampal subregions to link age-related impairments associated with discrimination of feature overlap and alternate response strategies to gene expression in specific hippocampal subregions.
Collapse
|
22
|
Yegla B, Foster T. Effect of Systemic Inflammation on Rat Attentional Function and Neuroinflammation: Possible Protective Role for Food Restriction. Front Aging Neurosci 2019; 11:296. [PMID: 31708767 PMCID: PMC6823289 DOI: 10.3389/fnagi.2019.00296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Aging is characterized by subtle cognitive decline, which correlates with increased peripheral inflammation. Acute activation of the peripheral immune system, via lipopolysaccharide (LPS) injection, elicits deficits in hippocampal-dependent spatial memory. Little is known concerning the effect of chronic inflammation on prefrontal cortex (PFC)-dependent vigilance. We examined the impact of repeated LPS injections in young and middle-age rats on the 5-choice serial reaction time task (5-CSRTT), expecting repeated LPS treatment to induce attentional deficits with greater disruption in middle-age. Methods: Male Fischer-344 rats, 4- and 12-months-old, were food restricted and trained on the 5-CSRTT. Once rats reached criterion, they were injected with LPS (1 mg/kg, i.p.) weekly for 4 weeks and testing started 48 h after each injection. To examine the possibility that mild food restriction inherent to the behavioral task influenced inflammation markers, a second group of food-restricted or ad-lib-fed rats was assessed for cytokine changes 48 h after one injection. Results: Performing LPS-treated rats exhibited a sickness response, manifesting as reduced initiated and completed trials during the first week but recovered by the second week of testing. After the first week, LPS-treated rats continued to exhibit longer response latencies, despite no change in food retrieval latency, suggestive of LPS-induced cognitive slowing. Similarly, LPS-induced impairment of attention was observed as increased omissions with heightened cognitive demand and increased age. Repeated LPS-treatment increased the level of PFC IL-1α, and PFC IL-6 was marginally higher in middle-age rats. No effect of age or treatment was observed for plasma cytokines in performing rats. Histological examination of microglia indicated increased colocalization of Iba1+ and CD68+ cells from middle-age relative to young rats. Examination of food restriction demonstrated an attenuation of age- and LPS-related increases in plasma cytokine levels. Conclusions: Systemic inflammation, induced through LPS treatment, impaired attentional function, which was independent of sickness and exacerbated by increased cognitive demand and increased age. Additional studies revealed that food restriction, associated with the task, attenuated markers of neuroinflammation and plasma cytokines. The results emphasize the need for improved methods for modeling low-level chronic systemic inflammation to effectively examine its impact on attention during aging.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Thomas Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
23
|
Hernandez AR, Hernandez CM, Truckenbrod LM, Campos KT, McQuail JA, Bizon JL, Burke SN. Age and Ketogenic Diet Have Dissociable Effects on Synapse-Related Gene Expression Between Hippocampal Subregions. Front Aging Neurosci 2019; 11:239. [PMID: 31607897 PMCID: PMC6755342 DOI: 10.3389/fnagi.2019.00239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/19/2019] [Indexed: 01/01/2023] Open
Abstract
As the number of individuals living beyond the age of 65 is rapidly increasing, so is the need to develop strategies to combat the age-related cognitive decline that may threaten independent living. Although the link between altered neuronal signaling and age-related cognitive impairments is not completely understood, it is evident that declining cognitive abilities are at least partially due to synaptic dysfunction. Aging is accompanied by well-documented changes in both excitatory and inhibitory synaptic signaling across species. Age-related synaptic alterations are not uniform across the brain, however, with different regions showing unique patterns of vulnerability in advanced age. In the hippocampus, increased activity within the CA3 subregion has been observed across species, and this can be reversed with anti-epileptic medication. In contrast to CA3, the dentate gyrus shows reduced activity with age and declining metabolic activity. Ketogenic diets have been shown to decrease seizure incidence and severity in epilepsy, improve metabolic function in diabetes type II, and improve cognitive function in aged rats. This link between neuronal activity and metabolism suggests that metabolic interventions may be able to ameliorate synaptic signaling deficits accompanying advanced age. We therefore investigated the ability of a dietary regimen capable of inducing nutritional ketosis and improving cognition to alter synapse-related gene expression across the dentate gyrus, CA3 and CA1 subregions of the hippocampus. Following 12 weeks of a ketogenic or calorie-matched standard diet, RTq-PCR was used to quantify expression levels of excitatory and inhibitory synaptic signaling genes within CA1, CA3 and dentate gyrus. While there were no age or diet-related changes in CA1 gene expression, expression levels were significantly altered within CA3 by age and within the dentate gyrus by diet for several genes involved in presynaptic glutamate regulation and postsynaptic excitation and plasticity. These data demonstrate subregion-specific alterations in synaptic signaling with age and the potential for a ketogenic diet to alter these processes in dissociable ways across different brain structures that are uniquely vulnerable in older animals.
Collapse
Affiliation(s)
- Abbi R. Hernandez
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
| | - Caesar M. Hernandez
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
| | - Leah M. Truckenbrod
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
| | - Keila T. Campos
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
| | - Joseph A. McQuail
- Department of Physiology, Pharmacology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Jennifer L. Bizon
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
| | - Sara N. Burke
- Department of Neurosciences, University of Florida, Gainesville, FL, United States
- Institute on Aging, University of Florida, Gainesville, FL, United States
| |
Collapse
|
24
|
Foster TC. Senescent neurophysiology: Ca 2+ signaling from the membrane to the nucleus. Neurobiol Learn Mem 2019; 164:107064. [PMID: 31394200 DOI: 10.1016/j.nlm.2019.107064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 12/16/2022]
Abstract
The current review provides a historical perspective on the evolution of hypothesized mechanisms for senescent neurophysiology, focused on the CA1 region of the hippocampus, and the relationship of senescent neurophysiology to impaired hippocampal-dependent memory. Senescent neurophysiology involves processes linked to calcium (Ca2+) signaling including an increase in the Ca2+-dependent afterhyperpolarization (AHP), decreasing pyramidal cell excitability, hyporesponsiveness of N-methyl-D-aspartate (NMDA) receptor function, and a shift in Ca2+-dependent synaptic plasticity. Dysregulation of intracellular Ca2+ and downstream signaling of kinase and phosphatase activity lies at the core of senescent neurophysiology. Ca2+-dysregulation involves a decrease in Ca2+ influx through NMDA receptors and an increase release of Ca2+ from internal Ca2+ stores. Recent work has identified changes in redox signaling, arising in middle-age, as an initiating factor for senescent neurophysiology. The shift in redox state links processes of aging, oxidative stress and inflammation, with functional changes in mechanisms required for episodic memory. The link between age-related changes in Ca2+ signaling, epigenetics and gene expression is an exciting area of research. Pharmacological and behavioral intervention, initiated in middle-age, can promote memory function by initiating transcription of neuroprotective genes and rejuvenating neurophysiology. However, with more advanced age, or under conditions of neurodegenerative disease, epigenetic changes may weaken the link between environmental influences and transcription, decreasing resilience of memory function.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience and Genetics and Genomics Program, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
25
|
Age and Sex Influence the Hippocampal Response and Recovery Following Sepsis. Mol Neurobiol 2019; 56:8557-8572. [PMID: 31278440 PMCID: PMC6834928 DOI: 10.1007/s12035-019-01681-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/14/2019] [Indexed: 01/03/2023]
Abstract
Although in-hospital mortality rates for sepsis have decreased, survivors often experience lasting physical and cognitive deficits. Moreover, older adults are more vulnerable to long-term complications associated with sepsis. We employed a murine model to examine the influence of age and sex on the brain’s response and recovery following sepsis. Young (~ 4 months) and old (~ 20 months) mice (C57BL/6) of both sexes underwent cecal ligation and puncture (CLP) with restraint stress. The hippocampal transcriptome was examined in age- and sex-matched controls at 1 and 4 days post-CLP. In general, immune- and stress-related genes increased, while neuronal, synaptic, and glial genes decreased 1 day after CLP-induced sepsis. However, specific age and sex differences were observed for the initial responsiveness to sepsis as well as the rate of recovery examined on day 4. Young females exhibited a muted transcriptional response relative to young males and old females. Old females exhibited a robust shift in gene transcription on day 1, and while most genes recovered, genes linked to neurogenesis and myelination continued to be downregulated by day 4. In contrast, old males exhibited a more delayed or prolonged response to sepsis, such that neuronal and synaptic genes continued to decrease while immune response genes continued to increase on day 4. These results suggest that aging is associated with delayed recovery from sepsis, which is particularly evident in males.
Collapse
|
26
|
Neuwirth LS, Masood S, Anderson DW, Schneider JS. The attention set-shifting test is sensitive for revealing sex-based impairments in executive functions following developmental lead exposure in rats. Behav Brain Res 2019; 366:126-134. [PMID: 30878351 PMCID: PMC6732195 DOI: 10.1016/j.bbr.2019.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/22/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022]
Abstract
The literature on lead (Pb) exposure has focused in large part on hippocampal-based learning and memory deficits, although frontoexecutive dysfunctions are known to exist in Pb-exposed humans. This study examined the effects of perinatal (PERI) and early postnatal (EPN) developmental low-level Pb-exposures in rats on frontoexecutive functions, using the Attention Set-Shift Test (ASST). Control males and females performed the ASST similarly. Male EPN rats had difficulty with simple discrimination (SD) of odors and failed to complete the compound discrimination (CD) stage of the ASST. All other Pb-exposed rats completed the training and testing. Male PERI rats performed worse on the SD, intradimensional (ID), and intradimensional-reversal (ID-Rev) ASST stages when compared to male Control rats. Female EPN rats performed similar to Controls on the ID-Rev rats, whereas PERI rats performed better the trials-to-criterion on the ID-Rev than EPN and Control rats. Pb-exposed female rats had significant difficulty performing the ED/ED-Rev stages, with the number of trials-to-criterion double that required by Pb-exposed and Control male rats and Control female rats. Together, the ASST results showed that developmental Pb-exposure induces frontoexecutive dysfunction that persists into adulthood, with different sex-based vulnerabilities dependent upon the time-period of neurotoxicant exposure.
Collapse
Affiliation(s)
- Lorenz S Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY 11568, United States; SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury NY 11568, United States; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| | - Sidrah Masood
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY 11568, United States; SUNY Neuroscience Research Institute, SUNY Old Westbury, Old Westbury NY 11568, United States
| | - David W Anderson
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Jay S Schneider
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| |
Collapse
|
27
|
Lupo G, Gaetani S, Cacci E, Biagioni S, Negri R. Molecular Signatures of the Aging Brain: Finding the Links Between Genes and Phenotypes. Neurotherapeutics 2019; 16:543-553. [PMID: 31161490 PMCID: PMC6694319 DOI: 10.1007/s13311-019-00743-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aging is associated with cognitive decline and increased vulnerability to neurodegenerative diseases. The progressive extension of the average human lifespan is bound to lead to a corresponding increase in the fraction of cognitively impaired elderly individuals among the human population, with an enormous societal and economic burden. At the cellular and tissue levels, cognitive decline is linked to a reduction in specific neuronal subpopulations, a widespread decrease in synaptic plasticity and an increase in neuroinflammation due to an enhanced activation of astrocytes and microglia, but the molecular mechanisms underlying these functional changes during normal aging and in neuropathological conditions remain poorly understood. In this review, we summarize very recent and outstanding progress in elucidating the molecular changes associated with cognitive decline through the genome-wide profiling of aging brain cells at different molecular levels (genomic, epigenomic, transcriptomic, proteomic). We discuss how the correlation of different molecular and phenotypic traits driven by mathematical and computational analyses of large datasets has led to the prediction of key molecular nodes of neurodegenerative pathways, and provide a few examples of candidate regulators of cognitive decline identified with these approaches. Furthermore, we highlight the dysregulation of the synaptic transcriptome in neuronal cells and of the inflammatory transcriptome in glial cells as some of the key events during normal and neuropathological human brain aging.
Collapse
Affiliation(s)
- Giuseppe Lupo
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro, 00185, Rome, Italy.
| | - Silvana Gaetani
- Department of Physiology and Farmacology "V. Erspamer", Sapienza University of Rome, Piazzale A. Moro, 00185, Rome, Italy
| | - Emanuele Cacci
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro, 00185, Rome, Italy
| | - Stefano Biagioni
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro, 00185, Rome, Italy
| | - Rodolfo Negri
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Piazzale A. Moro, 00185, Rome, Italy
| |
Collapse
|
28
|
Duggan MR, Joshi S, Tan YF, Slifker M, Ross EA, Wimmer M, Parikh V. Transcriptomic changes in the prefrontal cortex of rats as a function of age and cognitive engagement. Neurobiol Learn Mem 2019; 163:107035. [PMID: 31185277 DOI: 10.1016/j.nlm.2019.107035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/05/2019] [Accepted: 06/08/2019] [Indexed: 01/19/2023]
Abstract
Although changes in cognitive functions including attention are well documented in aging, the neurobiological basis for such alterations is not fully understood. Increasing evidence points towards the contribution of genetic factors in age-related cognitive decline. However, genetic studies have remained inconsistent in characterizing specific genes that could predict functional decline in aging. Here we utilized next generation RNA sequencing (RNA-seq) to identify patterns of differentially expressed genes in the prefrontal cortex (PFC), a brain region implicated in attention, of young and aged animals that were either cognitively trained or had limited cognitive engagement. Consistent with previous investigations, aging alone was associated with increased expression of genes involved in multiple facets of innate and adaptive immune responses. On the contrary, the expression of immunity-related transcripts was reduced by cognitive engagement. In addition, transcripts across a wide range of cellular processes, including those associated with neuronal remodeling and plasticity, were upregulated by this behavioral manipulation. Surprisingly, aged subjects accounted for higher mean counts of upregulated transcripts and lower mean counts for downregulated transcripts as compared to the young subjects. Because aged rats exhibited lower attentional capacities, it is plausible that transcriptional changes associated with performance in these animals were reflective of compensatory changes that occurred to cope with the declining integrity of PFC functioning. Interestingly, the effects of both aging and cognitive engagement resulted in an upregulation of transcripts linked to extracellular exosomes, suggesting such extracellular vesicles may moderate a reciprocal gene by environment interaction in order to facilitate the reorganization of PFC circuitry and maintain functionality. Taken together, these findings provide novel insights into the capacities of both cognitive engagement as well as aging to alter gene expression in the PFC, and how the effects of such dynamic factors relate to variation in age-related cognitive abilities.
Collapse
Affiliation(s)
- Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Surbhi Joshi
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Yin-Fei Tan
- Genetics Research Facilities, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| | - Michael Slifker
- Biostatisitics and Bionformatics Facilities, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| | - Eric A Ross
- Biostatisitics and Bionformatics Facilities, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| | - Mathieu Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| |
Collapse
|
29
|
Ryan L, Hay M, Huentelman MJ, Duarte A, Rundek T, Levin B, Soldan A, Pettigrew C, Mehl MR, Barnes CA. Precision Aging: Applying Precision Medicine to the Field of Cognitive Aging. Front Aging Neurosci 2019; 11:128. [PMID: 31231204 PMCID: PMC6568195 DOI: 10.3389/fnagi.2019.00128] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
The current "one size fits all" approach to our cognitive aging population is not adequate to close the gap between cognitive health span and lifespan. In this review article, we present a novel model for understanding, preventing, and treating age-related cognitive impairment (ARCI) based on concepts borrowed from precision medicine. We will discuss how multiple risk factors can be classified into risk categories because of their interrelatedness in real life, the genetic variants that increase sensitivity to, or ameliorate, risk for ARCI, and the brain drivers or common mechanisms mediating brain aging. Rather than providing a definitive model of risk for ARCI and cognitive decline, the Precision Aging model is meant as a starting point to guide future research. To that end, after briefly discussing key risk categories, genetic risks, and brain drivers, we conclude with a discussion of steps that must be taken to move the field forward.
Collapse
Affiliation(s)
- Lee Ryan
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Meredith Hay
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Matt J. Huentelman
- Neurobehavioral Research Unit, Division of Neurological Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Audrey Duarte
- Center for Advanced Brain Imaging, School of Psychology, Georgia Institute of Technology, Atlanta, GA, United States
| | - Tatjana Rundek
- Clinical and Translational Research Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Bonnie Levin
- Neuropsychology Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anja Soldan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Matthias R. Mehl
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Carol A. Barnes
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
30
|
Younis RM, Taylor RM, Beardsley PM, McClay JL. The ANKS1B gene and its associated phenotypes: focus on CNS drug response. Pharmacogenomics 2019; 20:669-684. [PMID: 31250731 PMCID: PMC6912848 DOI: 10.2217/pgs-2019-0015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
The ANKS1B gene was a top finding in genome-wide association studies (GWAS) of antipsychotic drug response. Subsequent GWAS findings for ANKS1B include cognitive ability, educational attainment, body mass index, response to corticosteroids and drug dependence. We review current human association evidence for ANKS1B, in addition to functional studies that include two published mouse knockouts. The several GWAS findings in humans indicate that phenotypically relevant variation is segregating at the ANKS1B locus. ANKS1B shows strong plausibility for involvement in CNS drug response because it encodes a postsynaptic effector protein that mediates long-term changes to neuronal biology. Forthcoming data from large biobanks should further delineate the role of ANKS1B in CNS drug response.
Collapse
Affiliation(s)
- Rabha M Younis
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| | - Rachel M Taylor
- Center for Military Psychiatry & Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MA 20910, USA
| | - Patrick M Beardsley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- Center for Biomarker Research & Personalized Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| |
Collapse
|
31
|
Harman MF, Martín MG. Epigenetic mechanisms related to cognitive decline during aging. J Neurosci Res 2019; 98:234-246. [PMID: 31045277 DOI: 10.1002/jnr.24436] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 04/04/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Cognitive decline is a hallmark of the aging nervous system, characterized by increasing memory loss and a deterioration of mental capacity, which in turn creates a favorable context for the development of neurodegenerative diseases. One of the most detrimental alterations that occur at the molecular level in the brain during aging is the modification of the epigenetic mechanisms that control gene expression. As a result of these epigenetic-driven changes in the transcriptome most of the functions of the brain including synaptic plasticity, learning, and memory decline with aging. The epigenetic mechanisms altered during aging include DNA methylation, histone modifications, nucleosome remodeling, and microRNA-mediated gene regulation. In this review, we examine the current evidence concerning the changes of epigenetic modifications together with the molecular mechanisms underlying impaired neuronal gene transcription during aging. Herein, we discuss the alterations of DNA methylation pattern that occur in old neurons. We will also describe the most prominent age-related histone posttranslational modifications in the brain since changes in acetylation and methylation of specific lysine residues on H3 and H4 are associated to functional decline in the old. In addition, we discuss the role that changes in the levels of certain miRNAs would play in cognitive decline with aging. Finally, we provide an overview about the mechanisms either extrinsic or intrinsic that would trigger epigenetic changes in the aging brain, and the consequences of these changes, i.e., altered transcriptional profile and reactivation of transposable elements in old brain.
Collapse
Affiliation(s)
- María F Harman
- Instituto Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina.,Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mauricio G Martín
- Instituto Ferreyra, INIMEC-CONICET-UNC, Córdoba, Argentina.,Facultad de Ciencias Exactas Físicas y Naturales, Cátedra de Química Orgánica, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
32
|
Burke SN, Foster TC. Animal models of cognitive aging and circuit-specific vulnerability. HANDBOOK OF CLINICAL NEUROLOGY 2019; 167:19-36. [PMID: 31753133 DOI: 10.1016/b978-0-12-804766-8.00002-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Medial temporal lobe and prefrontal cortical structures are particularly vulnerable to dysfunction in advanced age and neurodegenerative diseases. This review focuses on cognitive aging studies in animals to illustrate the important aspects of the animal model paradigm for investigation of age-related memory and executive function loss. Particular attention is paid to the discussion of the face, construct, and predictive validity of animal models for determining the possible mechanisms of regional vulnerability in aging and for identifying novel therapeutic strategies. Aging is associated with a host of regionally specific neurobiologic alterations. Thus, targeted interventions that restore normal activity in one brain region may exacerbate aberrant activity in another, hindering the restoration of function at the behavioral level. As such, interventions that target the optimization of "cognitive networks" rather than discrete brain regions may be more effective for improving functional outcomes in the elderly.
Collapse
Affiliation(s)
- Sara N Burke
- Department of Neuroscience, William L. and Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Thomas C Foster
- Department of Neuroscience, William L. and Evelyn F. McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
33
|
Kumar A, Rani A, Scheinert RB, Ormerod BK, Foster TC. Nonsteroidal anti-inflammatory drug, indomethacin improves spatial memory and NMDA receptor function in aged animals. Neurobiol Aging 2018; 70:184-193. [PMID: 30031231 DOI: 10.1016/j.neurobiolaging.2018.06.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/04/2018] [Accepted: 06/19/2018] [Indexed: 11/17/2022]
Abstract
A redox-mediated decrease in N-methyl-D-aspartate (NMDA) receptor function contributes to psychiatric diseases and impaired cognition during aging. Inflammation provides a potential source of reactive oxygen species for inducing NMDA receptor hypofunction. The present study tested the hypothesis that the nonsteroidal anti-inflammatory drug indomethacin, which improves spatial episodic memory in aging rats, would enhance NMDA receptor function through a shift in the redox state. Male F344 young and aged rats were prescreened using a 1-day version of the water maze task. Animals were then treated with the indomethacin or vehicle, delivered in a frozen milk treat (orally, twice per day, 18 days), and retested on the water maze. Indomethacin treatment enhanced water maze performance. Hippocampal slices were prepared for examination of CA3-CA1 synaptic responses, long-term potentiation, and NMDA receptor-mediated synaptic responses. No effect of treatment was observed for the total synaptic response. Long-term potentiation magnitude and NMDA receptor input-output curves were enhanced for aged indomethacin-treated animals. To examine redox regulation of NMDA receptors, a second group of aged animals was treated with indomethacin or vehicle, and the effect of the reducing agent, dithiothreitol ([DTT], 0.5 mM) on NMDA receptor-mediated synaptic responses was evaluated. As expected, DTT increased the NMDA receptor response and the effect of DTT was reduced by indomethacin treatment. The results indicate that indomethacin acted to diminish the age-related and redox-mediated NMDA receptor hypofunction and suggest that inflammation contributes to cognitive impairment through an increase in redox stress.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Rachel B Scheinert
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, J. Crayton Pruitt Family, University of Florida, Gainesville, FL, USA
| | - Brandi K Ormerod
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, J. Crayton Pruitt Family, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
34
|
Abstract
SIGNIFICANCE Oxidative stress increases in the brain with aging and neurodegenerative diseases. Previous work emphasized irreversible oxidative damage in relation to cognitive impairment. This research has evolved to consider a continuum of alterations, from redox signaling to oxidative damage, which provides a basis for understanding the onset and progression of cognitive impairment. This review provides an update on research linking redox signaling to altered function of neural circuits involved in information processing and memory. Recent Advances: Starting in middle age, redox signaling triggers changes in nervous system physiology described as senescent physiology. Hippocampal senescent physiology involves decreased cell excitability, altered synaptic plasticity, and decreased synaptic transmission. Recent studies indicate N-methyl-d-aspartate and ryanodine receptors and Ca2+ signaling molecules as molecular substrates of redox-mediated senescent physiology. CRITICAL ISSUES We review redox homeostasis mechanisms and consider the chemical character of reactive oxygen and nitrogen species and their role in regulating different transmitter systems. In this regard, senescent physiology may represent the co-opting of pathways normally responsible for feedback regulation of synaptic transmission. Furthermore, differences across transmitter systems may underlie differential vulnerability of brain regions and neuronal circuits to aging and disease. FUTURE DIRECTIONS It will be important to identify the intrinsic mechanisms for the shift in oxidative/reductive processes. Intrinsic mechanism will depend on the transmitter system, oxidative stressors, and expression/activity of antioxidant enzymes. In addition, it will be important to identify how intrinsic processes interact with other aging factors, including changes in inflammatory or hormonal signals. Antioxid. Redox Signal. 28, 1724-1745.
Collapse
Affiliation(s)
- Ashok Kumar
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Brittney Yegla
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Thomas C Foster
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida.,2 Genetics and Genomics Program, Genetics Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
35
|
Abstract
Gene expression in the aging brain depends on transcription signals generated by senescent physiology, interacting with genetic and epigenetic programs. In turn, environmental factors influence epigenetic mechanisms, such that an epigenetic-environmental link may contribute to the accumulation of cellular damage, susceptibility or resilience to stressors, and variability in the trajectory of age-related cognitive decline. Epigenetic mechanisms, DNA methylation and histone modifications, alter chromatin structure and the accessibility of DNA. Furthermore, small non-coding RNA, termed microRNA (miRNA) bind to messenger RNA (mRNA) to regulate translation. In this review, we examine key questions concerning epigenetic mechanisms in regulating the expression of genes associated with brain aging and age-related cognitive decline. In addition, we highlight the interaction of epigenetics with senescent physiology and environmental factors in regulating transcription.
Collapse
Affiliation(s)
- Jolie D Barter
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Thomas C Foster
- 1 Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,2 Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Mladenova D, Barry G, Konen LM, Pineda SS, Guennewig B, Avesson L, Zinn R, Schonrock N, Bitar M, Jonkhout N, Crumlish L, Kaczorowski DC, Gong A, Pinese M, Franco GR, Walkley CR, Vissel B, Mattick JS. Adar3 Is Involved in Learning and Memory in Mice. Front Neurosci 2018; 12:243. [PMID: 29719497 PMCID: PMC5914295 DOI: 10.3389/fnins.2018.00243] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/27/2018] [Indexed: 11/13/2022] Open
Abstract
The amount of regulatory RNA encoded in the genome and the extent of RNA editing by the post-transcriptional deamination of adenosine to inosine (A-I) have increased with developmental complexity and may be an important factor in the cognitive evolution of animals. The newest member of the A-I editing family of ADAR proteins, the vertebrate-specific ADAR3, is highly expressed in the brain, but its functional significance is unknown. In vitro studies have suggested that ADAR3 acts as a negative regulator of A-I RNA editing but the scope and underlying mechanisms are also unknown. Meta-analysis of published data indicates that mouse Adar3 expression is highest in the hippocampus, thalamus, amygdala, and olfactory region. Consistent with this, we show that mice lacking exon 3 of Adar3 (which encodes two double stranded RNA binding domains) have increased levels of anxiety and deficits in hippocampus-dependent short- and long-term memory formation. RNA sequencing revealed a dysregulation of genes involved in synaptic function in the hippocampi of Adar3-deficient mice. We also show that ADAR3 transiently translocates from the cytoplasm to the nucleus upon KCl-mediated activation in SH-SY5Y cells. These results indicate that ADAR3 contributes to cognitive processes in mammals.
Collapse
Affiliation(s)
- Dessislava Mladenova
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Guy Barry
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Lyndsey M. Konen
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- St. Vincent's Centre for Applied Medical Research (AMR), Sydney, NSW, Australia
| | - Sandy S. Pineda
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Boris Guennewig
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Lotta Avesson
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Raphael Zinn
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- St. Vincent's Centre for Applied Medical Research (AMR), Sydney, NSW, Australia
| | - Nicole Schonrock
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Maina Bitar
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Nicky Jonkhout
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Lauren Crumlish
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | | | - Andrew Gong
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Mark Pinese
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Gloria R. Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carl R. Walkley
- St. Vincent's Institute of Medical Research, Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Bryce Vissel
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- St. Vincent's Centre for Applied Medical Research (AMR), Sydney, NSW, Australia
| | - John S. Mattick
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
37
|
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: 4.0] [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
|
38
|
Ianov L, Riva A, Kumar A, Foster TC. DNA Methylation of Synaptic Genes in the Prefrontal Cortex Is Associated with Aging and Age-Related Cognitive Impairment. Front Aging Neurosci 2017; 9:249. [PMID: 28824413 PMCID: PMC5539085 DOI: 10.3389/fnagi.2017.00249] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/14/2017] [Indexed: 01/17/2023] Open
Abstract
The current study investigates DNA methylation as a possible epigenetic regulator of transcription associated with aging and cognitive function. Young and aged male Fischer 344 rats were behaviorally characterized on a set shifting task, and whole genome bisulfite sequencing was employed to profile the DNA methylome of the medial prefrontal cortex (mPFC). DNA methylation was also compared to RNA expression in the mPFC from the same animals. Variability in methylation was mainly observed for CpG sites as opposed to CHG and CHH sites. Gene bodies, specifically introns, contain the highest levels of methylation. During aging, hypermethylation was observed for genes linked to synaptic function and GTPase activity. Furthermore, impaired cognitive flexibility during aging was associated with hypermethylation of genes linked to postsynaptic density, dendrites, the axon terminus, and Ca2+ channels. Finally, comparison with RNA expression confirmed that hypermethylation was correlated with decreased expression of synaptic genes. The results indicate that DNA methylation over the lifespan contributes to synaptic modification observed in brain aging and age-related cognitive impairment.
Collapse
Affiliation(s)
- Lara Ianov
- Department of Neuroscience, McKnight Brain Institute, University of Florida, GainesvilleFL, United States.,Genetics and Genomics Program, Genetics Institute, University of Florida, GainesvilleFL, United States
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, GainesvilleFL, United States
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, GainesvilleFL, United States
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, GainesvilleFL, United States.,Genetics and Genomics Program, Genetics Institute, University of Florida, GainesvilleFL, United States
| |
Collapse
|
39
|
French L, Ma T, Oh H, Tseng GC, Sibille E. Age-Related Gene Expression in the Frontal Cortex Suggests Synaptic Function Changes in Specific Inhibitory Neuron Subtypes. Front Aging Neurosci 2017; 9:162. [PMID: 28611654 PMCID: PMC5446995 DOI: 10.3389/fnagi.2017.00162] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/09/2017] [Indexed: 12/22/2022] Open
Abstract
Genome-wide expression profiling of the human brain has revealed genes that are differentially expressed across the lifespan. Characterizing these genes adds to our understanding of both normal functions and pathological conditions. Additionally, the specific cell-types that contribute to the motor, sensory and cognitive declines during aging are unclear. Here we test if age-related genes show higher expression in specific neural cell types. Our study leverages data from two sources of murine single-cell expression data and two sources of age-associations from large gene expression studies of postmortem human brain. We used nonparametric gene set analysis to test for age-related enrichment of genes associated with specific cell-types; we also restricted our analyses to specific gene ontology groups. Our analyses focused on a primary pair of single-cell expression data from the mouse visual cortex and age-related human post-mortem gene expression information from the orbitofrontal cortex. Additional pairings that used data from the hippocampus, prefrontal cortex, somatosensory cortex and blood were used to validate and test specificity of our findings. We found robust age-related up-regulation of genes that are highly expressed in oligodendrocytes and astrocytes, while genes highly expressed in layer 2/3 glutamatergic neurons were down-regulated across age. Genes not specific to any neural cell type were also down-regulated, possibly due to the bulk tissue source of the age-related genes. A gene ontology-driven dissection of the cell-type enriched genes highlighted the strong down-regulation of genes involved in synaptic transmission and cell-cell signaling in the Somatostatin (Sst) neuron subtype that expresses the cyclin dependent kinase 6 (Cdk6) and in the vasoactive intestinal peptide (Vip) neuron subtype expressing myosin binding protein C, slow type (Mybpc1). These findings provide new insights into cell specific susceptibility to normal aging, and suggest age-related synaptic changes in specific inhibitory neuron subtypes.
Collapse
Affiliation(s)
- Leon French
- Neurobiology of Depression and Aging Lab, Centre for Addiction and Mental Health, Campbell Family Mental Health Research InstituteToronto, ON, Canada.,Department of Psychiatry, University of TorontoToronto, ON, Canada.,Institute of Medical Science, University of TorontoToronto, ON, Canada
| | - TianZhou Ma
- Department of Biostatistics, University of PittsburghPittsburgh, PA, United States
| | - Hyunjung Oh
- Neurobiology of Depression and Aging Lab, Centre for Addiction and Mental Health, Campbell Family Mental Health Research InstituteToronto, ON, Canada
| | - George C Tseng
- Department of Biostatistics, University of PittsburghPittsburgh, PA, United States
| | - Etienne Sibille
- Neurobiology of Depression and Aging Lab, Centre for Addiction and Mental Health, Campbell Family Mental Health Research InstituteToronto, ON, Canada.,Department of Psychiatry, University of TorontoToronto, ON, Canada.,Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
| |
Collapse
|