1
|
Kundu S, Amini M, Stachon T, Fries F, Seitz B, Li Z, Li S, Liu S, Shu SL, Suiwal S, Szentmáry N. Effect of isolated keratin 3 knockdown on gene expression of primary limbal epithelial cells without and with inflammatory stimuli. Ann Anat 2025; 260:152670. [PMID: 40334825 DOI: 10.1016/j.aanat.2025.152670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025]
Abstract
PURPOSE Studies have shown that keratin 3 (KRT3) expression is reduced in paired box 6 (PAX6) haploinsufficient primary limbal epithelial cells (LECs). The downregulation of KRT3 expression due to PAX6 haploinsufficiency is likely a critical factor in the development and progression of aniridia associated keratopathy (AAK). In addition, the ocular surface of congenital aniridia patients exhibits an inflammatory environment. The objective of this study was to investigate the isolated effect of KRT3 knockdown, achieved via siRNA silencing in healthy LECs, on PAX6 and other related gene expressions, both under normal and inflammatory conditions. METHODS To achieve KRT3 knockdown, human primary LECs were transfected with KRT3 siRNA using Lipofectamine 2000. Inflammatory conditions were induced 48 hours after transfection by treating the cells with 2 mg/mL of lipopolysaccharides (LPS) or 1 ng/mL of IL-1β. Subsequently, gene and protein expression levels were analysed using qPCR, Western blotting, and ELISA. RESULTS Following KRT3 knockdown at protein level, there was DSG1, ADH7 and PPARγ upregulation and MAPK1 downregulation solely at transcriptional level (p ≤ 0.031). Nevertheless, IL-6 downregulation could be observed both at transcriptional and at protein levels (p ≤ 0.003). Following KRT3 siRNA knockdown, LPS induced inflammation decreased PPARγ mRNA level and IL-1β induced inflammation decreased DSG1 and ADH7 mRNA levels without changes at protein levels (p ≤ 0.014). In contrast, in control knockdown LECs, IL-1β induced inflammation significantly decreased KRT3 mRNA and protein levels and IL-6 protein level (p ≤ 0.02). CONCLUSIONS In normal LECs, inflammatory stimuli slow differentiation and simultaneously induce IL-6 production. These mechanisms are absent in KRT3 knockdown LECs. As a result, despite the presence of inflammation, KRT3 knockdown LECs continue their differentiation unaltered while maintaining inflammatory IL-6 protein secretion.
Collapse
Affiliation(s)
- Swarnali Kundu
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany.
| | - Maryam Amini
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Tanja Stachon
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Fabian Fries
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany; Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany
| | - Zhen Li
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Shuailin Li
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Shanhe Liu
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Shao-Lun Shu
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Shweta Suiwal
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| | - Nóra Szentmáry
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Congenital Aniridia Research, Saarland University, Homburg, Saar, Germany
| |
Collapse
|
2
|
Hossain R, Noonong K, Nuinoon M, Majima HJ, Eawsakul K, Sompol P, Rahman MA, Tangpong J. Network Pharmacology, Molecular Docking, and In Vitro Insights into the Potential of Mitragyna speciosa for Alzheimer's Disease. Int J Mol Sci 2024; 25:13201. [PMID: 39684911 DOI: 10.3390/ijms252313201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Mitragyna speciosa Korth. Havil (MS) has a traditional use in relieving pain, managing hypertension, treating cough, and diarrhea, and as a morphine substitute in addiction recovery. Its potential in addressing Alzheimer's disease (AD), a neurodegenerative condition with no effective treatments, is under investigation. This study aims to explore MS mechanisms in treating AD through network pharmacology, molecular docking, and in vitro studies. Using network pharmacology, we identified 19 MS components that may affect 60 AD-related targets. The compound-target network highlighted significant interactions among 60 nodes and 470 edges, with an average node degree of 15.7. The KEGG enrichment analysis revealed Alzheimer's disease (hsa05010) as a relevant pathway. We connected 20 targets to tau and β-amyloid proteins through gene expression data from the AlzData database. Docking studies demonstrated high binding affinities of MS compounds like acetylursolic acid, beta-sitosterol, isomitraphylline, and speciophylline to AD-related proteins, such as AKT1, GSK3B, NFκB1, and BACE1. In vitro studies showed that ethanolic (EE), distilled water (DWE), and pressurized hot water (PHWE) extracts of MS-treated 100 μM H2O2-induced SH-SY5Y cells significantly reduced oxidative damage. This research underscores the multi-component, multi-target, and multi-pathway effects of MS on AD, providing insights for future research and potential clinical applications.
Collapse
Affiliation(s)
- Rahni Hossain
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Kunwadee Noonong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Manit Nuinoon
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
- Hematology and Transfusion Science Research Center, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Hideyuki J Majima
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Komgrit Eawsakul
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Pradoldej Sompol
- Department of Pharmacology & Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Jitbanjong Tangpong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Excellence Center for Innovation and Health Product (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
3
|
Forero SA, Liu S, Shetty N, Ophir AG. Re-wiring of the bonded brain: Gene expression among pair bonded female prairie voles changes as they transition to motherhood. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12906. [PMID: 38861664 PMCID: PMC11166254 DOI: 10.1111/gbb.12906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
Motherhood is a costly life-history transition accompanied by behavioral and neural plasticity necessary for offspring care. Motherhood in the monogamous prairie vole is associated with decreased pair bond strength, suggesting a trade-off between parental investment and pair bond maintenance. Neural mechanisms governing pair bonds and maternal bonds overlap, creating possible competition between the two. We measured mRNA expression of genes encoding receptors for oxytocin (oxtr), dopamine (d1r and d2r), mu-opioids (oprm1a), and kappa-opioids (oprk1a) within three brain areas processing salience of sociosensory cues (anterior cingulate cortex; ACC), pair bonding (nucleus accumbens; NAc), and maternal care (medial preoptic area; MPOA). We compared gene expression differences between pair bonded prairie voles that were never pregnant, pregnant (~day 16 of pregnancy), and recent mothers (day 3 of lactation). We found greater gene expression in the NAc (oxtr, d2r, oprm1a, and oprk1a) and MPOA (oxtr, d1r, d2r, oprm1a, and oprk1a) following the transition to motherhood. Expression for all five genes in the ACC was greatest for females that had been bonded for longer. Gene expression within each region was highly correlated, indicating that oxytocin, dopamine, and opioids comprise a complimentary gene network for social signaling. ACC-NAc gene expression correlations indicated that being a mother (oxtr and d1r) or maintaining long-term pair bonds (oprm1a) relies on the coordination of different signaling systems within the same circuit. Our study suggests the maternal brain undergoes changes that prepare females to face the trade-off associated with increased emotional investment in offspring, while also maintaining a pair bond.
Collapse
MESH Headings
- Animals
- Female
- Arvicolinae/genetics
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Pair Bond
- Maternal Behavior/physiology
- Nucleus Accumbens/metabolism
- Pregnancy
- Receptors, Oxytocin/genetics
- Receptors, Oxytocin/metabolism
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Gyrus Cinguli/metabolism
- Preoptic Area/metabolism
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
Collapse
Affiliation(s)
| | - Sydney Liu
- Department of PsychologyCornell UniversityIthacaNew YorkUSA
| | - Netra Shetty
- Department of PsychologyCornell UniversityIthacaNew YorkUSA
| | | |
Collapse
|
4
|
Fernández-Albarral JA, Ramírez AI, de Hoz R, Matamoros JA, Salobrar-García E, Elvira-Hurtado L, López-Cuenca I, Sánchez-Puebla L, Salazar JJ, Ramírez JM. Glaucoma: from pathogenic mechanisms to retinal glial cell response to damage. Front Cell Neurosci 2024; 18:1354569. [PMID: 38333055 PMCID: PMC10850296 DOI: 10.3389/fncel.2024.1354569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Glaucoma is a neurodegenerative disease of the retina characterized by the irreversible loss of retinal ganglion cells (RGCs) leading to visual loss. Degeneration of RGCs and loss of their axons, as well as damage and remodeling of the lamina cribrosa are the main events in the pathogenesis of glaucoma. Different molecular pathways are involved in RGC death, which are triggered and exacerbated as a consequence of a number of risk factors such as elevated intraocular pressure (IOP), age, ocular biomechanics, or low ocular perfusion pressure. Increased IOP is one of the most important risk factors associated with this pathology and the only one for which treatment is currently available, nevertheless, on many cases the progression of the disease continues, despite IOP control. Thus, the IOP elevation is not the only trigger of glaucomatous damage, showing the evidence that other factors can induce RGCs death in this pathology, would be involved in the advance of glaucomatous neurodegeneration. The underlying mechanisms driving the neurodegenerative process in glaucoma include ischemia/hypoxia, mitochondrial dysfunction, oxidative stress and neuroinflammation. In glaucoma, like as other neurodegenerative disorders, the immune system is involved and immunoregulation is conducted mainly by glial cells, microglia, astrocytes, and Müller cells. The increase in IOP produces the activation of glial cells in the retinal tissue. Chronic activation of glial cells in glaucoma may provoke a proinflammatory state at the retinal level inducing blood retinal barrier disruption and RGCs death. The modulation of the immune response in glaucoma as well as the activation of glial cells constitute an interesting new approach in the treatment of glaucoma.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lidia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Juan J. Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
5
|
Liharska L, Charney A. Transcriptomics : Approaches to Quantifying Gene Expression and Their Application to Studying the Human Brain. Curr Top Behav Neurosci 2024; 68:129-176. [PMID: 38972894 DOI: 10.1007/7854_2024_466] [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] [Indexed: 07/09/2024]
Abstract
To date, the field of transcriptomics has been characterized by rapid methods development and technological advancement, with new technologies continuously rendering older ones obsolete.This chapter traces the evolution of approaches to quantifying gene expression and provides an overall view of the current state of the field of transcriptomics, its applications to the study of the human brain, and its place in the broader emerging multiomics landscape.
Collapse
Affiliation(s)
- Lora Liharska
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | | |
Collapse
|
6
|
Li Y, Li R, Wang N, Gu J, Gao J. Gender effects on autism spectrum disorder: a multi-site resting-state functional magnetic resonance imaging study of transcriptome-neuroimaging. Front Neurosci 2023; 17:1203690. [PMID: 37409103 PMCID: PMC10318192 DOI: 10.3389/fnins.2023.1203690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction The gender disparity in autism spectrum disorder (ASD) has been one of the salient features of condition. However, its relationship between the pathogenesis and genetic transcription in patients of different genders has yet to reach a reliable conclusion. Methods To address this gap, this study aimed to establish a reliable potential neuro-marker in gender-specific patients, by employing multi-site functional magnetic resonance imaging (fMRI) data, and to further investigate the role of genetic transcription molecules in neurogenetic abnormalities and gender differences in autism at the neuro-transcriptional level. To this end, age was firstly used as a regression covariate, followed by the use of ComBat to remove the site effect from the fMRI data, and abnormal functional activity was subsequently identified. The resulting abnormal functional activity was then correlated by genetic transcription to explore underlying molecular functions and cellular molecular mechanisms. Results Abnormal brain functional activities were identified in autism patients of different genders, mainly located in the default model network (DMN) and precuneus-cingulate gyrus-frontal lobe. The correlation analysis of neuroimaging and genetic transcription further found that heterogeneous brain regions were highly correlated with genes involved in signal transmission between neurons' plasma membranes. Additionally, we further identified different weighted gene expression patterns and specific expression tissues of risk genes in ASD of different genders. Discussion Thus, this work not only identified the mechanism of abnormal brain functional activities caused by gender differences in ASD, but also explored the genetic and molecular characteristics caused by these related changes. Moreover, we further analyzed the genetic basis of sex differences in ASD from a neuro-transcriptional perspective.
Collapse
Affiliation(s)
- Yanling Li
- School of Electrical Engineering and Electronic Information, Xihua University, Chengdu, China
| | - Rui Li
- School of Electrical Engineering and Electronic Information, Xihua University, Chengdu, China
| | - Ning Wang
- School of Electrical Engineering and Electronic Information, Xihua University, Chengdu, China
| | - Jiahe Gu
- School of Electrical Engineering and Electronic Information, Xihua University, Chengdu, China
| | - Jingjing Gao
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Makowski C, Wang H, Srinivasan A, Qi A, Qiu Y, van der Meer D, Frei O, Zou J, Visscher P, Yang J, Chen CH. Larger cerebral cortex is genetically correlated with greater frontal area and dorsal thickness. Proc Natl Acad Sci U S A 2023; 120:e2214834120. [PMID: 36893272 PMCID: PMC10089183 DOI: 10.1073/pnas.2214834120] [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: 08/30/2022] [Accepted: 01/18/2023] [Indexed: 03/11/2023] Open
Abstract
Human cortical expansion has occurred non-uniformly across the brain. We assessed the genetic architecture of cortical global expansion and regionalization by comparing two sets of genome-wide association studies of 24 cortical regions with and without adjustment for global measures (i.e., total surface area, mean cortical thickness) using a genetically informed parcellation in 32,488 adults. We found 393 and 756 significant loci with and without adjusting for globals, respectively, where 8% and 45% loci were associated with more than one region. Results from analyses without adjustment for globals recovered loci associated with global measures. Genetic factors that contribute to total surface area of the cortex particularly expand anterior/frontal regions, whereas those contributing to thicker cortex predominantly increase dorsal/frontal-parietal thickness. Interactome-based analyses revealed significant genetic overlap of global and dorsolateral prefrontal modules, enriched for neurodevelopmental and immune system pathways. Consideration of global measures is important in understanding the genetic variants underlying cortical morphology.
Collapse
Affiliation(s)
- Carolina Makowski
- Department of Radiology, University of California San Diego, La Jolla, CA92093
| | - Hao Wang
- Department of Radiology, University of California San Diego, La Jolla, CA92093
| | - Anjali Srinivasan
- Department of Radiology, University of California San Diego, La Jolla, CA92093
| | - Anna Qi
- Department of Radiology, University of California San Diego, La Jolla, CA92093
| | - Yuqi Qiu
- School of Statistics, East China Normal University, Shanghai20050, China
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research Centre, Division of Mental Health and Addiction, University of Oslo, Oslo0450, Norway
| | - Oleksandr Frei
- Norwegian Centre for Mental Disorders Research Centre, Division of Mental Health and Addiction, University of Oslo, Oslo0450, Norway
| | - Jingjing Zou
- Division of Biostatistics and Bioinformatics, University of California San Diego, La Jolla, CA92093
| | - Peter M. Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
| | - Jian Yang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang310024, China
| | - Chi-Hua Chen
- Department of Radiology, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
8
|
Ng PY, McNeely TL, Baker DJ. Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J 2023; 290:1326-1339. [PMID: 34873840 PMCID: PMC9167891 DOI: 10.1111/febs.16315] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Microglial homeostasis has emerged as a critical mediator of health and disease in the central nervous system. In their neuroprotective role as the predominant immune cells of the brain, microglia surveil the microenvironment for debris and pathogens, while also promoting neurogenesis and performing maintenance on synapses. Chronological ageing, disease onset, or traumatic injury promotes irreparable damage or deregulated signaling to reinforce neurotoxic phenotypes in microglia. These insults may include cellular senescence, a stable growth arrest often accompanied by the production of a distinctive pro-inflammatory secretory phenotype, which may contribute to age- or disease-driven decline in neuronal health and cognition and is a potential novel therapeutic target. Despite this increased scrutiny, unanswered questions remain about what distinguishes senescent microglia and non-senescent microglia reacting to insults occurring in ageing, disease, and injury, and how central the development of senescence is in their pivot from guardian to assailant. To intelligently design future studies to untangle senescent microglia from other primed and reactionary states, specific criteria must be developed that define this population and allow for comparisons between different model systems. Comparing microglial activity seen in homeostasis, ageing, disease, and injury allows for a more coherent understanding of when and how senescent and other harmful microglial subpopulations should be targeted.
Collapse
Affiliation(s)
- Pei Y Ng
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Taylor L McNeely
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
9
|
Jiang WR, Wu W, Yang LJ, Yang W, Tian Q, Yao ZH. Alteration of Cognitive Function in Aging and Alzheimer's Disease Mice Is Related to Dysfunction of the Neuroimmune System. J Alzheimers Dis 2023; 94:815-839. [PMID: 37334607 DOI: 10.3233/jad-230292] [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] [Indexed: 06/20/2023]
Abstract
BACKGROUND Both Alzheimer's disease (AD) and aging have aging-related cognitive dysfunction with a high incidence. These neurological diseases cause serious cognitive problems in patients' daily life. But the cognitive dysfunction mechanism in-depth of aging is far less known than that of AD. OBJECTIVE To reveal the different mechanisms of AD and aging-related cognitive dysfunction, we compared the mechanisms of aging and AD through analysis of differentially expressed genes. METHODS Mice were divided into four groups (3-month C57BL, 16-month C57BL, 3-month 3xTg AD mice, and 16-month 3xTg AD mice) according to genotype and age. The Morris water maze was employed to investigate the spatial cognition of mice. Differential expressions of genes of AD and aging were analyzed through RNA sequencing and GO, KEGG, Reactome analysis, and the dynamic change trend analysis. Microglia was stained with immunofluorescence and its numbers were counted for analysis. RESULTS The cognitive function of elderly mice were worse through testing with the Morris water maze. The cognitive function of 16-month 3xTg AD mice were worse than 16-month C57BL mice. The alteration tendencies of DE genes were uncovered, and microglia numbers increased during aging and AD progression through immunofluorescence. CONCLUSION These results suggest that immune-related pathways might play a critical role in aging and AD-related cognitive dysfunction. Our research will help to provide some new potential targets for treating cognitive dysfunction in aging and AD.
Collapse
Affiliation(s)
- Wan-Rong Jiang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li-Jie Yang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wanzhexi Yang
- Department of Physiology, Pharmacology and Neuroscience, University College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Yao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Mamelak M. The Treatment of Parkinson's Disease with Sodium Oxybate. Curr Mol Pharmacol 2023; 16:564-579. [PMID: 36330625 DOI: 10.2174/1874467216666221103121135] [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: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Sodiun Oxybate (SO) has a number of attributes that may mitigate the metabolic stress on the substantia nigra pars compacta (SNpc) dopaminergic (DA) neurons in Parkinson's disease (PD). These neurons function at the borderline of energy sufficiency. SO is metabolized to succinate and supplies energy to the cell by generating ATP. SO is a GABAB agonist and, as such, also arrests the high energy requiring calcium pace-making activity of these neurons. In addition, blocking calcium entry impedes the synaptic release and subsequent neurotransmission of aggregated synuclein species. As DA neurons degenerate, a homeostatic failure exposes these neurons to glutamate excitotoxicity, which in turn accelerates the damage. SO inhibits the neuronal release of glutamate and blocks its agonistic actions. Most important, SO generates NADPH, the cell's major antioxidant cofactor. Excessive free radical production within DA neurons and even more so within activated microglia are early and key features of the degenerative process that are present long before the onset of motor symptoms. NADPH maintains cell glutathione levels and alleviates oxidative stress and its toxic consequences. SO, a histone deacetylase inhibitor also suppresses the expression of microglial NADPH oxidase, the major source of free radicals in Parkinson brain. The acute clinical use of SO at night has been shown to reduce daytime sleepiness and fatigue in patients with PD. With long-term use, its capacity to supply energy to DA neurons, impede synuclein transmission, block excitotoxicity and maintain an anti-oxidative redox environment throughout the night may delay the onset of PD and slow its progress.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Moca EN, Lecca D, Hope KT, Etienne F, Schaler AW, Espinoza K, Chappell MS, Gray DT, Tweedie D, Sidhu S, Masukawa L, Sitoy H, Mathew R, Saban DR, Greig NH, De Biase LM. Microglia Drive Pockets of Neuroinflammation in Middle Age. J Neurosci 2022; 42:3896-3918. [PMID: 35396327 PMCID: PMC9097782 DOI: 10.1523/jneurosci.1922-21.2022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/18/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
During aging, microglia produce inflammatory factors, show reduced tissue surveillance, altered interactions with synapses, and prolonged responses to CNS insults, positioning these cells to have profound impact on the function of nearby neurons. We and others recently showed that microglial attributes differ significantly across brain regions in young adult mice. However, the degree to which microglial properties vary during aging is largely unexplored. Here, we analyze and manipulate microglial aging within the basal ganglia, brain circuits that exhibit prominent regional microglial heterogeneity and where neurons are vulnerable to functional decline and neurodegenerative disease. In male and female mice, we demonstrate that VTA and SNc microglia exhibit unique and premature responses to aging, compared with cortex and NAc microglia. This is associated with localized VTA/SNc neuroinflammation that may compromise synaptic function as early as middle age. Surprisingly, systemic inflammation, local neuron death, and astrocyte aging do not appear to underlie these early aging responses of VTA and SNc microglia. Instead, we found that microglial lysosome status was tightly linked to early aging of VTA microglia. Microglial ablation/repopulation normalized VTA microglial lysosome swelling and suppressed increases in VTA microglial density during aging. In contrast, CX3CR1 receptor KO exacerbated VTA microglial lysosome rearrangements and VTA microglial proliferation during aging. Our findings reveal a previously unappreciated regional variation in onset and magnitude of microglial proliferation and inflammatory factor production during aging and highlight critical links between microglial lysosome status and local microglial responses to aging.SIGNIFICANCE STATEMENT Microglia are CNS cells that are equipped to regulate neuronal health and function throughout the lifespan. We reveal that microglia in select brain regions begin to proliferate and produce inflammatory factors in late middle age, months before microglia in other brain regions. These findings demonstrate that CNS neuroinflammation during aging is not uniform. Moreover, they raise the possibility that local microglial responses to aging play a critical role in determining which populations of neurons are most vulnerable to functional decline and neurodegenerative disease.
Collapse
Affiliation(s)
- Eric N Moca
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Daniela Lecca
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland 21224
| | - Keenan T Hope
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Fanny Etienne
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Ari W Schaler
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Katherine Espinoza
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Megan S Chappell
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Daniel T Gray
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - David Tweedie
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland 21224
| | - Shanaya Sidhu
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Lindsay Masukawa
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Hannah Sitoy
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Rose Mathew
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Daniel R Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Nigel H Greig
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland 21224
| | - Lindsay M De Biase
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| |
Collapse
|
12
|
Joo YH, Lee MW, Son YD, Chang KA, Yaqub M, Kim HK, Cumming P, Kim JH. In Vivo Cerebral Translocator Protein (TSPO) Binding and Its Relationship with Blood Adiponectin Levels in Treatment-Naïve Young Adults with Major Depression: A [ 11C]PK11195 PET Study. Biomedicines 2021; 10:biomedicines10010034. [PMID: 35052718 PMCID: PMC8773340 DOI: 10.3390/biomedicines10010034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/05/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Adiponectin is an adipokine that mediates cellular cholesterol efflux and plays important roles in neuroinflammatory processes. In this study, we undertook positron emission tomography (PET) with the translocator protein (TSPO) ligand [11C]PK11195 and measured serum adiponectin levels in groups of treatment-naïve young adult patients with major depressive disorder (MDD) and matched healthy controls. Thirty treatment-naïve MDD patients (median age: 24 years) and twenty-three healthy controls underwent [11C]PK11195 PET. We quantified TSPO availability in brain as the [11C]PK11195 binding potential (BPND) using a reference tissue model in conjunction with the supervised cluster analysis (SVCA4) algorithm. Age, sex distribution, body mass index, and serum adiponectin levels did not differ between the groups. Between-group analysis using a region-of-interest approach showed significantly higher [11C]PK11195 BPND in the left anterior and right posterior cingulate cortices in MDD patients than in controls. Serum adiponectin levels had significant negative correlations with [11C]PK11195 BPND in the bilateral hippocampus in MDD patients, but significant positive correlations in the bilateral hippocampus in the control group. Our results indicate significantly higher TSPO binding in the anterior and posterior cingulate cortices in treatment-naïve young MDD patients, suggesting microglial activation in these limbic regions, which are involved in cognitive and emotional processing. The opposite correlations between [11C]PK11195 BPND in the hippocampus with serum adiponectin levels in MDD and control groups suggest that microglial activation in the hippocampus may respond differentially to adiponectin signaling in MDD and healthy subjects, possibly with respect to microglial phenotype.
Collapse
Affiliation(s)
- Yo-Han Joo
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
| | - Min-Woo Lee
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
| | - Young-Don Son
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Department of Pharmacology, Gachon University College of Medicine, Gachon University, Incheon 21936, Korea
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands;
| | - Hang-Keun Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon 21936, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, CH-3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane 4059, Australia
| | - Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea; (Y.-H.J.); (M.-W.L.); (Y.-D.S.); (K.-A.C.); (H.-K.K.)
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Gachon University, Incheon 21565, Korea
- Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Gachon University, Incheon 21565, Korea
- Correspondence: ; Tel.: +82-32-460-2696
| |
Collapse
|
13
|
Mavrikaki M, Lee JD, Solomon IH, Slack FJ. Severe COVID-19 induces molecular signatures of aging in the human brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.11.24.21266779. [PMID: 34845457 PMCID: PMC8629201 DOI: 10.1101/2021.11.24.21266779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is predominantly an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and remains a significant threat to public health. COVID-19 is accompanied by neurological symptoms and cognitive decline, but the molecular mechanisms underlying this effect remain unclear. As aging induces distinct molecular signatures in the brain associated with cognitive decline in healthy populations, we hypothesized that COVID-19 may induce molecular signatures of aging. Here, we performed whole transcriptomic analysis of human frontal cortex, a critical area for cognitive function, in 12 COVID-19 cases and age- and sex-matched uninfected controls. COVID-19 induces profound changes in gene expression, despite the absence of detectable virus in brain tissue. Pathway analysis shows downregulation of genes involved in synaptic function and cognition and upregulation of genes involved in immune processes. Comparison with five independent transcriptomic datasets of aging human frontal cortex reveals striking similarities between aged individuals and severe COVID-19 patients. Critically, individuals below 65 years of age exhibit profound transcriptomic changes not observed among older individuals in our patient cohort. Our data indicate that severe COVID-19 induces molecular signatures of aging in the human brain and emphasize the value of neurological follow-up in recovered individuals.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- These authors contributed equally: Maria Mavrikaki and Jonathan D. Lee
- Correspondence to M.M. and F.J.S.: ;
| | - Jonathan D. Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- These authors contributed equally: Maria Mavrikaki and Jonathan D. Lee
| | - Isaac H. Solomon
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02215, USA
- Correspondence to M.M. and F.J.S.: ;
| |
Collapse
|
14
|
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: 7] [Impact Index Per Article: 1.8] [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
|
15
|
Kazemi T, Huang S, Avci NG, Akay YM, Akay M. Investigating the effects of chronic perinatal alcohol and combined nicotine and alcohol exposure on dopaminergic and non-dopaminergic neurons in the VTA. Sci Rep 2021; 11:8706. [PMID: 33888815 PMCID: PMC8062589 DOI: 10.1038/s41598-021-88221-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
16
|
Neurotrophins Time Point Intervention after Traumatic Brain Injury: From Zebrafish to Human. Int J Mol Sci 2021; 22:ijms22041585. [PMID: 33557335 PMCID: PMC7915547 DOI: 10.3390/ijms22041585] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) remains the leading cause of long-term disability, which annually involves millions of individuals. Several studies on mammals reported that neurotrophins could play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and TBI. This protective role of neurotrophins after an event of TBI has also been reported in the zebrafish model. Nevertheless, reparative mechanisms in mammalian brain are limited, and newly formed neurons do not survive for a long time. In contrast, the brain of adult fish has high regenerative properties after brain injury. The evident differences in regenerative properties between mammalian and fish brain have been ascribed to remarkable different adult neurogenesis processes. However, it is not clear if the specific role and time point contribution of each neurotrophin and receptor after TBI is conserved during vertebrate evolution. Therefore, in this review, I reported the specific role and time point of intervention for each neurotrophic factor and receptor after an event of TBI in zebrafish and mammals.
Collapse
|
17
|
Yang L, Li S, Mo C, Zhou B, Fan S, Shi F, Wei X, Zhao Q, Yang G, Li S, Mou C. Transcriptome analysis and identification of age-associated fertility decreased genes in hen uterovaginal junction. Poult Sci 2020; 100:100892. [PMID: 33516476 PMCID: PMC7936153 DOI: 10.1016/j.psj.2020.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 11/17/2020] [Accepted: 12/01/2020] [Indexed: 11/05/2022] Open
Abstract
Avian sperm storage tubules (SSTs), which are located in the uterovaginal junction (UVJ) of the oviduct, are primary sperm storage sites after mating or artificial insemination. The mechanism underlying reduced sperm storage efficiency of SSTs which is highly correlated with decreased fertility rates in aged laying breeders remains largely unclear. Here, comparative transcriptomic analysis between the aged and young White Leghorn hens (120 vs. 30 wk) was applied to identify gene expression changes of UVJs containing SSTs. Bioinformatics analysis revealed 567 upregulated and 1998 downregulated differentially expressed genes. Gene ontology analysis was highly enriched in terms of immune system, cell adhesion, and cytoskeleton proteins. Kyoto Encyclopedia of Genes and Genomes analysis revealed 5 significant (P < 0.05) pathways including inositol phosphate and glycerophospholipid metabolism. β-Galactosidase staining of chicken UVJ sections suggested increased cell senescence via aging. Oil Red O staining and immunohistochemistry detection of ADFP both confirmed distribution of lipid droplets in SST cells with increased intensity in aged breeders. The lipid synthesis and metabolism-related genes represented by TFAP2 and PLD1 were differentially expressed in aged laying breeders. The upregulation of IL15 and downregulation of a large number of immune-related genes in aged breeders indicate altered immune homeostasis in UVJs and SSTs. The increased accumulation of lipids, and altered immunity homeostasis, combined with other factors (TJP1, MYL9, AFDN, and RPL13, etc.) are potentially dominant effectors to decrease the sperm storage efficiency and egg fertility in aged laying breeders.
Collapse
Affiliation(s)
- Liubin Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Shaomei Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Changhuan Mo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Baogui Zhou
- Department of Poultry Breeding, Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 100000 China
| | - Shijie Fan
- Department of Poultry Breeding, Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 100000 China
| | - Fengying Shi
- Department of Poultry Breeding, Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 100000 China
| | - Xiaoran Wei
- Department of Poultry Breeding, Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 100000 China
| | - Qianqian Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Ge Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Shijun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China
| | - Chunyan Mou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070 China.
| |
Collapse
|
18
|
Stonebarger GA, Urbanski HF, Woltjer RL, Vaughan KL, Ingram DK, Schultz PL, Calderazzo SM, Siedeman JA, Mattison JA, Rosene DL, Kohama SG. Amyloidosis increase is not attenuated by long-term calorie restriction or related to neuron density in the prefrontal cortex of extremely aged rhesus macaques. GeroScience 2020; 42:1733-1749. [PMID: 32876855 PMCID: PMC7732935 DOI: 10.1007/s11357-020-00259-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/24/2020] [Indexed: 01/30/2023] Open
Abstract
As human lifespan increases and the population ages, diseases of aging such as Alzheimer's disease (AD) are a major cause for concern. Although calorie restriction (CR) as an intervention has been shown to increase healthspan in many species, few studies have examined the effects of CR on brain aging in primates. Using postmortem tissue from a cohort of extremely aged rhesus monkeys (22-44 years old, average age 31.8 years) from a longitudinal CR study, we measured immunohistochemically labeled amyloid beta plaques in Brodmann areas 32 and 46 of the prefrontal cortex, areas that play key roles in cognitive processing, are sensitive to aging and, in humans, are also susceptible to AD pathogenesis. We also evaluated these areas for cortical neuron loss, which has not been observed in younger cohorts of aged monkeys. We found a significant increase in plaque density with age, but this was unaffected by diet. Moreover, there was no change in neuron density with age or treatment. These data suggest that even in the oldest-old rhesus macaques, amyloid beta plaques do not lead to overt neuron loss. Hence, the rhesus macaque serves as a pragmatic animal model for normative human aging but is not a complete model of the neurodegeneration of AD. This model of aging may instead prove most useful for determining how even the oldest monkeys are protected from AD, and this information may therefore yield valuable information for clinical AD treatments.
Collapse
Affiliation(s)
- G A Stonebarger
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - H F Urbanski
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - R L Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - K L Vaughan
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Dickerson, MD, 20842, USA
- Charles River, Wilmington, MA, 01867, USA
| | - D K Ingram
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - P L Schultz
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - S M Calderazzo
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - J A Siedeman
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - J A Mattison
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, NIH, Dickerson, MD, 20842, USA
| | - D L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MD, 02218, USA
| | - S G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR, 97006, USA.
| |
Collapse
|
19
|
Advances in transcriptome analysis of human brain aging. Exp Mol Med 2020; 52:1787-1797. [PMID: 33244150 PMCID: PMC8080664 DOI: 10.1038/s12276-020-00522-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with gradual deterioration of physiological and biochemical functions, including cognitive decline. Transcriptome profiling of brain samples from individuals of varying ages has identified the whole-transcriptome changes that underlie age-associated cognitive declines. In this review, we discuss transcriptome-based research on human brain aging performed by using microarray and RNA sequencing analyses. Overall, decreased synaptic function and increased immune function are prevalent in most regions of the aged brain. Age-associated gene expression changes are also cell dependent and region dependent and are affected by genotype. In addition, the transcriptome changes that occur during brain aging include different splicing events, intersample heterogeneity, and altered levels of various types of noncoding RNAs. Establishing transcriptome-based hallmarks of human brain aging will improve the understanding of cognitive aging and neurodegenerative diseases and eventually lead to interventions that delay or prevent brain aging.
Collapse
|
20
|
Tong J, Williams B, Rusjan PM, Mizrahi R, Lacapère JJ, McCluskey T, Furukawa Y, Guttman M, Ang LC, Boileau I, Meyer JH, Kish SJ. Concentration, distribution, and influence of aging on the 18 kDa translocator protein in human brain: Implications for brain imaging studies. J Cereb Blood Flow Metab 2020; 40:1061-1076. [PMID: 31220997 PMCID: PMC7181090 DOI: 10.1177/0271678x19858003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Positron emission tomography (PET) imaging of the translocator protein (TSPO) is widely used as a biomarker of microglial activation. However, TSPO protein concentration in human brain has not been optimally quantified nor has its regional distribution been compared to TSPO binding. We determined TSPO protein concentration, change with age, and regional distribution by quantitative immunoblotting in autopsied human brain. Brain TSPO protein concentration (>0.1 ng/µg protein) was higher than those reported by in vitro binding assays by at least 2 to 70 fold. TSPO protein distributed widely in both gray and white matter regions, with distribution in major gray matter areas ranked generally similar to that of PET binding in second-generation radiotracer studies. TSPO protein concentration in frontal cortex was high at birth, declined precipitously during the first three months, and increased modestly during adulthood/senescence (10%/decade; vs. 30% for comparison astrocytic marker GFAP). As expected, TSPO protein levels were significantly increased (+114%) in degenerating putamen in multiple system atrophy, providing further circumstantial support for TSPO as a gliosis marker. Overall, findings show some similarities between TSPO protein and PET binding characteristics in the human brain but also suggest that part of the TSPO protein pool might be less available for radioligand binding.
Collapse
Affiliation(s)
- Junchao Tong
- Preclinical Imaging, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Junchao Tong, Preclinical Imaging, Centre
for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8,
Canada.
| | - Belinda Williams
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Pablo M. Rusjan
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Romina Mizrahi
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Jean-Jacques Lacapère
- Sorbonne Universités-UPMC University of
Paris 06, Département de Chimie, École Normale Supérieure-PSL Research University,
Paris, France
| | - Tina McCluskey
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Yoshiaki Furukawa
- Department of Neurology, Juntendo Tokyo
Koto Geriatric Medical Center, and Faculty of Medicine, University & Post
Graduate University of Juntendo, Tokyo, Japan
| | - Mark Guttman
- Centre for Movement Disorders, Toronto,
Ontario, Canada
| | - Lee-Cyn Ang
- Division of Neuropathology, London
Health Science Centre, University of Western Ontario, London, Ontario, Canada
| | - Isabelle Boileau
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
- Addiction Imaging Research Group,
Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario,
Canada
| | - Jeffrey H Meyer
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| | - Stephen J Kish
- Human Brain Laboratory, Research Imaging
Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Research Imaging Centre and Campbell
Family Mental Health Research Institute, Centre for Addiction and Mental Health,
Toronto, Ontario, Canada
| |
Collapse
|
21
|
Bondy SC. Aspects of the immune system that impact brain function. J Neuroimmunol 2020; 340:577167. [PMID: 32000018 DOI: 10.1016/j.jneuroim.2020.577167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
The conditions required for effective immune responses to viral or bacterial organisms and chemicals of exogenous origin and to intrinsic molecules of abnormal configuration, are briefly outlined. This is followed by a discussion of endocrine and environmental factors that can lead to excessive continuation of immune activity and persistent elevation of inflammatory responses. Such disproportionate activity becomes increasingly pronounced with aging and some possible reasons for this are considered. The specific vulnerability of the nervous system to prolonged immune events is involved in several disorders frequently found in the aging brain. In addition of being a target for inflammation associated with neurodegenerative disease, the nervous system is also seriously impacted by systemically widespread immune disturbances since there are several means by which immune information can access the CNS. The activation of glial cells and cells of non-nervous origin that form the basis of immune responses within the brain, can occur in differing modes resulting in widely differing consequences. The events underlying the relatively frequent occurrence of derangement and hyperreactivity of the immune system are considered, and a few potential ways of addressing this common condition are described.
Collapse
Affiliation(s)
- Stephen C Bondy
- Center for Occupational and Environmental Health, Department of Medicine, School of Medicine, University of California, Irvine, CA 92617-1830, USA.
| |
Collapse
|
22
|
Zhong Q, Zou Y, Liu H, Chen T, Zheng F, Huang Y, Chen C, Zhang Z. Toll-like receptor 4 deficiency ameliorates β2-microglobulin induced age-related cognition decline due to neuroinflammation in mice. Mol Brain 2020; 13:20. [PMID: 32059688 PMCID: PMC7023753 DOI: 10.1186/s13041-020-0559-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is a crucial receptor in neuroinflammation and apoptotic neuronal death, and increasing evidences indicated that β2-microglobulin (B2M) is thought to be a major contributor to age-related cognitive decline. In present study, we designed to investigate the effects of TLR4 on B2M-induced age-related cognitive decline. Wild-type (WT) C57BL/6, TLR4 knockout (TLR4 -KO) mice and hippocampal neurons from the two type mice were respectively divided into two groups: (1) Veh group; (2) B2M-treated group. The behavioral responses of mice were measured using Morris Water Maze. Hippocampal neurogenesis and neuronal damage, inflammatory response, apoptosis, synaptic proteins and neurotrophic factors, and TLR4/MyD88/NF-κB signaling pathway proteins were examined using molecular biological or histopathological methods. The results showed that WT mice received B2M in the DG exhibited age-related cognitive declines, increased TLR4 mRNA expression and high levels of interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α) and apoptotic neuronal death in the hippocampus, which were partially attenuated in TLR4-KO mice. Moreover, in absence of TLR4, B2M treatment improved hippocampus neurogenesis and increased synaptic related proteins. Our cell experiments further demonstrated that deletion of TLR4 could significantly increase synaptic related protein, decrease neuroinflammatory fators, inhibited apoptotic neuronal death, and regulated MyD88/NF-κB signal pathway after B2M treatment. In summary, our results support the TLR4 contributes to B2M-induced age-related cognitive decline due to neuroinflammation and apoptosis through TLR4/MyD88/NF-κB signaling pathway via a modulation of hippocampal neurogenesis and synaptic function. This may provide an important neuroprotective mechanism for improving age-related cognitive decline.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yufeng Zou
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Hongchao Liu
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
- Department of Anesthesiology, Maternal and Child Hospital of Hubei Province, Wuluo Road, Wuhan, 430071, Hubei, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Feng Zheng
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yifei Huang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Chang Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
23
|
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
|
24
|
Pöyhönen S, Er S, Domanskyi A, Airavaara M. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Front Physiol 2019; 10:486. [PMID: 31105589 PMCID: PMC6499070 DOI: 10.3389/fphys.2019.00486] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/08/2019] [Indexed: 12/28/2022] Open
Abstract
Astrocytes, oligodendrocytes, and microglia are abundant cell types found in the central nervous system and have been shown to play crucial roles in regulating both normal and disease states. An increasing amount of evidence points to the critical importance of glia in mediating neurodegeneration in Alzheimer’s and Parkinson’s diseases (AD, PD), and in ischemic stroke, where microglia are involved in initial tissue clearance, and astrocytes in the subsequent formation of a glial scar. The importance of these cells for neuronal survival has previously been studied in co-culture experiments and the search for neurotrophic factors (NTFs) initiated after finding that the addition of conditioned media from astrocyte cultures could support the survival of primary neurons in vitro. This led to the discovery of the potent dopamine neurotrophic factor, glial cell line-derived neurotrophic factor (GDNF). In this review, we focus on the relationship between glia and NTFs including neurotrophins, GDNF-family ligands, CNTF family, and CDNF/MANF-family proteins. We describe their expression in astrocytes, oligodendrocytes and their precursors (NG2-positive cells, OPCs), and microglia during development and in the adult brain. Furthermore, we review existing data on the glial phenotypes of NTF knockout mice and follow NTF expression patterns and their effects on glia in disease models such as AD, PD, stroke, and retinal degeneration.
Collapse
Affiliation(s)
- Suvi Pöyhönen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Safak Er
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Morin A, Mouzon B, Ferguson S, Paris D, Saltiel N, Lungmus C, Mullan M, Crawford F. Treatment With Nilvadipine Mitigates Inflammatory Pathology and Improves Spatial Memory in Aged hTau Mice After Repetitive Mild TBI. Front Aging Neurosci 2018; 10:292. [PMID: 30364309 PMCID: PMC6193195 DOI: 10.3389/fnagi.2018.00292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is the most common form of brain trauma worldwide. The effects of mTBI are not well-studied within the elderly population, yet older adults constitute a significant portion of all mTBI patients. Few preclinical studies have focused on the effects of mTBI, or mTBI treatments, in the aged brain, and none have explored repetitive mTBI (r-mTBI). In this study, we have administered our well-characterized 5-injury model (5 r-mTBI) to hTau mice aged 24 months to explore the neurobehavioral and neuropathological outcomes, and the effects of treatment with the dihydropyridine, Nilvadipine. Our previous studies have shown that Nilvadipine inhibits spleen tyrosine kinase (Syk), is effective at reducing inflammation, tau hyperphosphorylation, and amyloid production, and it has recently been investigated in a European Phase III clinical trial for Alzheimer’s disease (AD). In our 24-month-old r-mTBI mice, we observed increased neuroinflammation and a trend toward impaired cognitive performance compared to sham controls. Treatment with Nilvadipine mitigated the TBI-induced inflammatory response in aged r-mTBI animals and significantly improved spatial memory. To our knowledge, this is the only preclinical study focusing on the treatment of r-mTBI in aged, and these results suggest a therapeutic potential of Nilvadipine for consequences of mTBI.
Collapse
Affiliation(s)
- Alexander Morin
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Scott Ferguson
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Daniel Paris
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| | - Nicole Saltiel
- The Roskamp Institute, Sarasota, FL, United States.,James A Haley Veterans Administration, Tampa, FL, United States
| | | | - Mike Mullan
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, United States.,The Open University, Milton-Keynes, United Kingdom.,James A Haley Veterans Administration, Tampa, FL, United States
| |
Collapse
|
26
|
Kabba JA, Xu Y, Christian H, Ruan W, Chenai K, Xiang Y, Zhang L, Saavedra JM, Pang T. Microglia: Housekeeper of the Central Nervous System. Cell Mol Neurobiol 2018; 38:53-71. [PMID: 28534246 PMCID: PMC11481884 DOI: 10.1007/s10571-017-0504-2] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/16/2017] [Indexed: 12/17/2022]
Abstract
Microglia, of myeloid origin, play fundamental roles in the control of immune responses and the maintenance of central nervous system homeostasis. These cells, just like peripheral macrophages, may be activated into M1 pro-inflammatory or M2 anti-inflammatory phenotypes by appropriate stimuli. Microglia do not respond in isolation, but form part of complex networks of cells influencing each other. This review addresses the complex interaction of microglia with each cell type in the brain: neurons, astrocytes, cerebrovascular endothelial cells, and oligodendrocytes. We also highlight the participation of microglia in the maintenance of homeostasis in the brain, and their roles in the development and progression of age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- John Alimamy Kabba
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Yazhou Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Handson Christian
- Department of Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wenchen Ruan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Kitchen Chenai
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yun Xiang
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, People's Republic of China
| | - Luyong Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC, 20057, USA.
| |
Collapse
|
27
|
Ghassabian A, Sundaram R, Chahal N, McLain AC, Bell E, Lawrence DA, Yeung EH. Determinants of neonatal brain-derived neurotrophic factor and association with child development. Dev Psychopathol 2017; 29:1499-1511. [PMID: 28462726 PMCID: PMC6201316 DOI: 10.1017/s0954579417000414] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Using a population-based birth cohort in upstate New York (2008-2010), we examined the determinants of brain-derived neurotrophic factor (BDNF) measured in newborn dried blood spots (n = 2,637). We also examined the association between neonatal BDNF and children's development. The cohort was initially designed to examine the influence of infertility treatment on child development but found no impact. Mothers rated children's development in five domains repeatedly through age 3 years. Socioeconomic and maternal lifestyle determinants of BDNF were examined using multivariable linear regression models. Generalized linear mixed models estimated odds ratios for neonatal BDNF in relation to failing a developmental domain. Smoking and drinking in pregnancy, nulliparity, non-White ethnicity/race, and prepregnancy obesity were associated with lower neonatal BDNF. Neonatal BDNF was not associated with failure for developmental domains; however, there was an interaction between BDNF and preterm birth. In preterm infants, a higher BDNF was associated with lower odds of failing any developmental domains, after adjusting for confounders and infertility treatment. This result was particularly significant for failure in communication. Our findings suggest that BDNF levels in neonates may be impacted by maternal lifestyle characteristics. More specifically, lower neonatal BDNF might be an early marker of aberrant neurodevelopment in preterm infants.
Collapse
|
28
|
Shetty GA, Hattiangady B, Upadhya D, Bates A, Attaluri S, Shuai B, Kodali M, Shetty AK. Chronic Oxidative Stress, Mitochondrial Dysfunction, Nrf2 Activation and Inflammation in the Hippocampus Accompany Heightened Systemic Inflammation and Oxidative Stress in an Animal Model of Gulf War Illness. Front Mol Neurosci 2017; 10:182. [PMID: 28659758 PMCID: PMC5469946 DOI: 10.3389/fnmol.2017.00182] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/26/2017] [Indexed: 01/21/2023] Open
Abstract
Memory and mood dysfunction are the key symptoms of Gulf war illness (GWI), a lingering multi-symptom ailment afflicting >200,000 veterans who served in the Persian Gulf War-1. Research probing the source of the disease has demonstrated that concomitant exposures to anti-nerve gas agent pyridostigmine bromide (PB), pesticides, and war-related stress are among the chief causes of GWI. Indeed, exposures to GWI-related chemicals (GWIR-Cs) and mild stress in animal models cause memory and mood impairments alongside reduced neurogenesis and chronic low-level inflammation in the hippocampus. In the current study, we examined whether exposure to GWIR-Cs and stress causes chronic changes in the expression of genes related to increased oxidative stress, mitochondrial dysfunction, and inflammation in the hippocampus. We also investigated whether GWI is linked with chronically increased activation of Nrf2 (a master regulator of antioxidant response) in the hippocampus, and inflammation and enhanced oxidative stress at the systemic level. Adult male rats were exposed daily to low-doses of PB and pesticides (DEET and permethrin), in combination with 5 min of restraint stress for 4 weeks. Analysis of the hippocampus performed 6 months after the exposure revealed increased expression of many genes related to oxidative stress response and/or antioxidant activity (Hmox1, Sepp1, and Srxn1), reactive oxygen species metabolism (Fmo2, Sod2, and Ucp2) and oxygen transport (Ift172 and Slc38a1). Furthermore, multiple genes relevant to mitochondrial respiration (Atp6a1, Cox6a1, Cox7a2L, Ndufs7, Ndufv1, Lhpp, Slc25a10, and Ucp1) and neuroinflammation (Nfkb1, Bcl6, Csf2, IL6, Mapk1, Mapk3, Ngf, N-pac, and Prkaca) were up-regulated, alongside 73–88% reduction in the expression of anti-inflammatory genes IL4 and IL10, and nuclear translocation and increased expression of Nrf2 protein. These hippocampal changes were associated with elevated levels of pro-inflammatory cytokines and chemokines (Tnfa, IL1b, IL1a, Tgfb, and Fgf2) and lipid peroxidation byproduct malondialdehyde in the serum, suggesting the presence of an incessant systemic inflammation and elevated oxidative stress. These results imply that chronic oxidative stress, inflammation, and mitochondrial dysfunction in the hippocampus, and heightened systemic inflammation and oxidative stress likely underlie the persistent memory and mood dysfunction observed in GWI.
Collapse
Affiliation(s)
- Geetha A Shetty
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Bharathi Hattiangady
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Dinesh Upadhya
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Adrian Bates
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Bing Shuai
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Maheedhar Kodali
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Ashok K Shetty
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| |
Collapse
|
29
|
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
|
30
|
High BMI levels associate with reduced mRNA expression of IL10 and increased mRNA expression of iNOS (NOS2) in human frontal cortex. Transl Psychiatry 2017; 7:e1044. [PMID: 28244985 PMCID: PMC5545642 DOI: 10.1038/tp.2016.259] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/16/2016] [Accepted: 10/31/2016] [Indexed: 12/22/2022] Open
Abstract
Several studies link increasing body mass index (BMI) to cognitive decline both as a consequence of obesity per se and as a sequela of obesity-induced type 2 diabetes. Obese individuals are prone to a chronic low-grade inflammation as the metabolically active visceral fat produces proinflammatory cytokines. Animal studies indicate that these cytokines can cross the blood-brain barrier. Such crossover could potentially affect the immune system in the brain by inducing gene expression of proinflammatory genes. The relationship between obesity and neuroinflammation in the human brain is currently unknown. Therefore we aim to examine the relationship between BMI and gene expression of central inflammatory markers in the human frontal cortex. Microarray data of 141 neurologically and psychiatrically healthy individuals were obtained through the BrainCloud database. A simple linear regression analysis was performed with BMI as variable on data on IL10, IL1β, IL6, PTGS2 (COX2) and NOS2 (iNOS). Increasing BMI is associated with a decrease in the mRNA expression of IL10 (P=0.014) and an increase in the expression of NOS2 (iNOS; P=0.040). Expressions of IL10 and NOS2 (iNOS) were negatively correlated (P<0.001). The expression of IL10 was mostly affected by individuals with BMI ⩾40. Multiple linear regression analyses with BMI, age, sex and race as variables were performed in order to identify potential confounders. In conclusion, increasing BMI could affect the IL10-mediated anti-inflammatory defense in the brain and induce iNOS-mediated inflammatory activity.
Collapse
|
31
|
Niraula A, Sheridan JF, Godbout JP. Microglia Priming with Aging and Stress. Neuropsychopharmacology 2017; 42:318-333. [PMID: 27604565 PMCID: PMC5143497 DOI: 10.1038/npp.2016.185] [Citation(s) in RCA: 284] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/22/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023]
Abstract
The population of aged individuals is increasing worldwide and this has significant health and socio-economic implications. Clinical and experimental studies on aging have discovered myriad changes in the brain, including reduced neurogenesis, increased synaptic aberrations, higher metabolic stress, and augmented inflammation. In rodent models of aging, these alterations are associated with cognitive decline, neurobehavioral deficits, and increased reactivity to immune challenges. In rodents, caloric restriction and young blood-induced revitalization reverses the behavioral effects of aging. The increased inflammation in the aged brain is attributed, in part, to the resident population of microglia. For example, microglia of the aged brain are marked by dystrophic morphology, elevated expression of inflammatory markers, and diminished expression of neuroprotective factors. Importantly, the heightened inflammatory profile of microglia in aging is associated with a 'sensitized' or 'primed' phenotype. Mounting evidence points to a causal link between the primed profile of the aged brain and vulnerability to secondary insults, including infections and psychological stress. Conversely, psychological stress may also induce aging-like sensitization of microglia and increase reactivity to secondary challenges. This review delves into the characteristics of neuroinflammatory signaling and microglial sensitization in aging, its implications in psychological stress, and interventions that reverse aging-associated deficits.
Collapse
Affiliation(s)
- Anzela Niraula
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Division of Biosciences, The Ohio State University, College of Dentistry, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, 231 IBMR Bld, 460 Medical Center Drive Columbus, OH 43210, USA, Tel: +614 293 3456, Fax: +614 366 2097, E-mail:
| |
Collapse
|
32
|
Lubecka K, Kurzava L, Flower K, Buvala H, Zhang H, Teegarden D, Camarillo I, Suderman M, Kuang S, Andrisani O, Flanagan JM, Stefanska B. Stilbenoids remodel the DNA methylation patterns in breast cancer cells and inhibit oncogenic NOTCH signaling through epigenetic regulation of MAML2 transcriptional activity. Carcinogenesis 2016; 37:656-68. [PMID: 27207652 PMCID: PMC4936385 DOI: 10.1093/carcin/bgw048] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 03/20/2016] [Accepted: 04/15/2016] [Indexed: 12/30/2022] Open
Abstract
DNA hypomethylation was previously implicated in cancer progression and metastasis. The purpose of this study was to examine whether stilbenoids, resveratrol and pterostilbene thought to exert anticancer effects, target genes with oncogenic function for de novo methylation and silencing, leading to inactivation of related signaling pathways. Following Illumina 450K, genome-wide DNA methylation analysis reveals that stilbenoids alter DNA methylation patterns in breast cancer cells. On average, 75% of differentially methylated genes have increased methylation, and these genes are enriched for oncogenic functions, including NOTCH signaling pathway. MAML2, a coactivator of NOTCH targets, is methylated at the enhancer region and transcriptionally silenced in response to stilbenoids, possibly explaining the downregulation of NOTCH target genes. The increased DNA methylation at MAML2 enhancer coincides with increased occupancy of repressive histone marks and decrease in activating marks. This condensed chromatin structure is associated with binding of DNMT3B and decreased occupancy of OCT1 transcription factor at MAML2 enhancer, suggesting a role of DNMT3B in increasing methylation of MAML2 after stilbenoid treatment. Our results deliver a novel insight into epigenetic regulation of oncogenic signals in cancer and provide support for epigenetic-targeting strategies as an effective anticancer approach.
Collapse
Affiliation(s)
- Katarzyna Lubecka
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Lucinda Kurzava
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Kirsty Flower
- Epigenetic Unit, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Hannah Buvala
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | - Hao Zhang
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA
| | - Dorothy Teegarden
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Ignacio Camarillo
- Purdue University Center for Cancer Research, West Lafayette, IN, USA Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthew Suderman
- School of Social and Community Medicine, University of Bristol, Bristol, UK MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Shihuan Kuang
- Purdue University Center for Cancer Research, West Lafayette, IN, USA Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Ourania Andrisani
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - James M Flanagan
- Epigenetic Unit, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Barbara Stefanska
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA Purdue University Center for Cancer Research, West Lafayette, IN, USA
| |
Collapse
|
33
|
Dietary Linoleic Acid Lowering Reduces Lipopolysaccharide-Induced Increase in Brain Arachidonic Acid Metabolism. Mol Neurobiol 2016; 54:4303-4315. [PMID: 27339880 DOI: 10.1007/s12035-016-9968-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/08/2016] [Indexed: 12/29/2022]
Abstract
Linoleic acid (LA, 18:2n-6) is a precursor to arachidonic acid (AA, 20:4n-6), which can be converted by brain lipoxygenase and cyclooxygenase (COX) enzymes into various lipid mediators involved in the regulation of brain immunity. Brain AA metabolism is activated in rodents by the bacterial endotoxin, lipopolysaccharide (LPS). This study tested the hypothesis that dietary LA lowering, which limits plasma supply of AA to the brain, reduces LPS-induced upregulation in brain AA metabolism. Male Fischer CDF344 rats fed an adequate LA (5.2 % energy (en)) or low LA (0.4 % en) diet for 15 weeks were infused with LPS (250 ng/h) or vehicle into the fourth ventricle for 2 days using a mini-osmotic pump. The incorporation rate of intravenously infused unesterified 14C-AA into brain lipids, eicosanoids, and activities of phospholipase A2 and COX-1 and 2 enzymes were measured. Dietary LA lowering reduced the LPS-induced increase in prostaglandin E2 concentration and COX-2 activity (P < 0.05 by two-way ANOVA) without altering phospholipase activity. The 14C-AA incorporation rate into brain lipids was decreased by dietary LA lowering (P < 0.05 by two-way ANOVA). The present findings suggest that dietary LA lowering reduced LPS-induced increase in brain markers of AA metabolism. The clinical utility of LA lowering in brain disorders should be explored in future studies.
Collapse
|
34
|
Ianov L, Rani A, Beas BS, Kumar A, Foster TC. Transcription Profile of Aging and Cognition-Related Genes in the Medial Prefrontal Cortex. Front Aging Neurosci 2016; 8:113. [PMID: 27242522 PMCID: PMC4868850 DOI: 10.3389/fnagi.2016.00113] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/29/2016] [Indexed: 12/22/2022] Open
Abstract
Cognitive function depends on transcription; however, there is little information linking altered gene expression to impaired prefrontal cortex function during aging. Young and aged F344 rats were characterized on attentional set shift and spatial memory tasks. Transcriptional differences associated with age and cognition were examined using RNA sequencing to construct transcriptomic profiles for the medial prefrontal cortex (mPFC), white matter, and region CA1 of the hippocampus. The results indicate regional differences in vulnerability to aging. Age-related gene expression in the mPFC was similar to, though less robust than, changes in the dorsolateral PFC of aging humans suggesting that aging processes may be similar. Importantly, the pattern of transcription associated with aging did not predict cognitive decline. Rather, increased mPFC expression of genes involved in regulation of transcription, including transcription factors that regulate the strength of excitatory and inhibitory inputs, and neural activity-related immediate-early genes was observed in aged animals that exhibit delayed set shift behavior. The specificity of impairment on a mPFC-dependent task, associated with a particular mPFC transcriptional profile indicates that impaired executive function involves altered transcriptional regulation and neural activity/plasticity processes that are distinct from that described for impaired hippocampal function.
Collapse
Affiliation(s)
- Lara Ianov
- Department of Neuroscience, McKnight Brain Institute, University of FloridaGainesville, FL, USA
- Genetics and Genomics Program, Genetics Institute, University of FloridaGainesville, FL, USA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of FloridaGainesville, FL, USA
| | - Blanca S. Beas
- Department of Neuroscience, McKnight Brain Institute, University of FloridaGainesville, FL, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of FloridaGainesville, FL, USA
| | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain Institute, University of FloridaGainesville, FL, USA
- Genetics and Genomics Program, Genetics Institute, University of FloridaGainesville, FL, USA
| |
Collapse
|
35
|
De Filippis L, Halikere A, McGowan H, Moore JC, Tischfield JA, Hart RP, Pang ZP. Ethanol-mediated activation of the NLRP3 inflammasome in iPS cells and iPS cells-derived neural progenitor cells. Mol Brain 2016; 9:51. [PMID: 27160314 PMCID: PMC4862119 DOI: 10.1186/s13041-016-0221-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/08/2016] [Indexed: 12/19/2022] Open
Abstract
Background Alcohol abuse produces an enormous impact on health, society, and the economy. Currently, there are very limited therapies available, largely due to the poor understanding of mechanisms underlying alcohol use disorders (AUDs) in humans. Oxidative damage of mitochondria and cellular proteins aggravates the progression of neuroinflammation and neurological disorders initiated by alcohol abuse. Results Here we show that ethanol exposure causes neuroinflammation in both human induced pluripotent stem (iPS) cells and human neural progenitor cells (NPCs). Ethanol exposure for 24 hours or 7 days does not affect the proliferation of iPS cells and NPCs, but primes an innate immune-like response by activating the NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. This leads to an increase of microtubule-associated protein 1A/1B-light chain 3+ (LC3B+) autophagic puncta and impairment of the mitochondrial and lysosomal distribution. In addition, a decrease of mature neurons derived from differentiating NPCs is evident in ethanol pre-exposed compared to control NPCs. Moreover, a second insult of a pro-inflammatory factor in addition to ethanol preexposure enhances innate cellular inflammation in human iPS cells. Conclusions This study provides strong evidence that neuronal inflammation contributes to the pathophysiology of AUDs through the activation of the inflammasome pathway in human cellular models.
Collapse
Affiliation(s)
- Lidia De Filippis
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA. .,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.
| | - Apoorva Halikere
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Heather McGowan
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Jennifer C Moore
- Department of Genetics, Rutgers University, Piscataway, 08854, USA.,Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA
| | - Jay A Tischfield
- Department of Genetics, Rutgers University, Piscataway, 08854, USA.,Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA
| | - Ronald P Hart
- Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA.,Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, 08854, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA. .,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The review aims to provide a summary of recent developments in the study of gene expression in the aging human brain. RECENT FINDINGS Profiling differentially expressed genes or 'transcripts' in the human brain over the course of normal aging has provided valuable insights into the biological pathways that appear activated or suppressed in late life. Genes mediating neuroinflammation and immune system activation in particular, show significant age-related upregulation creating a state of vulnerability to neurodegenerative and neuropsychiatric disease in the aging brain. Cellular ionic dyshomeostasis and age-related decline in a host of molecular influences on synaptic efficacy may underlie neurocognitive decline in later life. Critically, these investigations have also shed light on the mobilization of protective genetic responses within the aging human brain that help determine health and disease trajectories in older age. There is growing interest in the study of pre and posttranscriptional regulators of gene expression, and the role of noncoding RNAs in particular, as mediators of the phenotypic diversity that characterizes human brain aging. SUMMARY Gene expression studies in healthy brain aging offer an opportunity to unravel the intricately regulated cellular underpinnings of neurocognitive aging as well as disease risk and resiliency in late life. In doing so, new avenues for early intervention in age-related neurodegenerative disease could be investigated with potentially significant implications for the development of disease-modifying therapies.
Collapse
|
37
|
Ramanan VK, Risacher SL, Nho K, Kim S, Shen L, McDonald BC, Yoder KK, Hutchins GD, West JD, Tallman EF, Gao S, Foroud TM, Farlow MR, De Jager PL, Bennett DA, Aisen PS, Petersen RC, Jack CR, Toga AW, Green RC, Jagust WJ, Weiner MW, Saykin AJ, for the Alzheimer’s Disease Neuroimaging Initiative (ADNI). GWAS of longitudinal amyloid accumulation on 18F-florbetapir PET in Alzheimer's disease implicates microglial activation gene IL1RAP. Brain 2015; 138:3076-88. [PMID: 26268530 PMCID: PMC4671479 DOI: 10.1093/brain/awv231] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/24/2015] [Indexed: 12/30/2022] Open
Abstract
Brain amyloid deposition is thought to be a seminal event in Alzheimer's disease. To identify genes influencing Alzheimer's disease pathogenesis, we performed a genome-wide association study of longitudinal change in brain amyloid burden measured by (18)F-florbetapir PET. A novel association with higher rates of amyloid accumulation independent from APOE (apolipoprotein E) ε4 status was identified in IL1RAP (interleukin-1 receptor accessory protein; rs12053868-G; P = 1.38 × 10(-9)) and was validated by deep sequencing. IL1RAP rs12053868-G carriers were more likely to progress from mild cognitive impairment to Alzheimer's disease and exhibited greater longitudinal temporal cortex atrophy on MRI. In independent cohorts rs12053868-G was associated with accelerated cognitive decline and lower cortical (11)C-PBR28 PET signal, a marker of microglial activation. These results suggest a crucial role of activated microglia in limiting amyloid accumulation and nominate the IL-1/IL1RAP pathway as a potential target for modulating this process.
Collapse
Affiliation(s)
- Vijay K Ramanan
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shannon L. Risacher
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kwangsik Nho
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,5 Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sungeun Kim
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,5 Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,5 Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brenna C. McDonald
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,6 Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karmen K. Yoder
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gary D. Hutchins
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John D. West
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eileen F. Tallman
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sujuan Gao
- 4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,7 Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tatiana M. Foroud
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,5 Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Martin R. Farlow
- 4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,6 Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Philip L. De Jager
- 8 Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Brigham and Women’s Hospital, Boston, MA 02115, USA,9 Departments of Neurology and Psychiatry, Harvard Medical School, Boston, MA 02115, USA,10 Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - David A. Bennett
- 11 Rush Alzheimer’s Disease Centre, Rush University Medical Centre, Chicago, IL 60612, USA
| | - Paul S. Aisen
- 12 University of Southern California Alzheimer's Therapeutic Research Institute, San Diego, CA 92121, USA
| | - Ronald C. Petersen
- 13 Department of Neurology, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | - Clifford R. Jack
- 14 Department of Radiology, Mayo Clinic Minnesota, Rochester, MN 55905, USA
| | - Arthur W. Toga
- 15 Laboratory of NeuroImaging, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Robert C. Green
- 16 Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - William J. Jagust
- 17 Department of Neurology, University of California, Berkeley, CA 94720, USA
| | - Michael W. Weiner
- 18 Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, CA 94143, USA,19 Department of Veterans Affairs Medical Centre, San Francisco, CA 94121, USA
| | - Andrew J. Saykin
- 1 Centre for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA,4 Indiana Alzheimer Disease Centre, Indiana University School of Medicine, Indianapolis, IN 46202, USA,5 Centre for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
38
|
Rapoport SI, Primiani CT, Chen CT, Ahn K, Ryan VH. Coordinated Expression of Phosphoinositide Metabolic Genes during Development and Aging of Human Dorsolateral Prefrontal Cortex. PLoS One 2015; 10:e0132675. [PMID: 26168237 PMCID: PMC4500567 DOI: 10.1371/journal.pone.0132675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/17/2015] [Indexed: 01/10/2023] Open
Abstract
Background Phosphoinositides, lipid-signaling molecules, participate in diverse brain processes within a wide metabolic cascade. Hypothesis Gene transcriptional networks coordinately regulate the phosphoinositide cascade during human brain Development and Aging. Methods We used the public BrainCloud database for human dorsolateral prefrontal cortex to examine age-related expression levels of 49 phosphoinositide metabolic genes during Development (0 to 20+ years) and Aging (21+ years). Results We identified three groups of partially overlapping genes in each of the two intervals, with similar intergroup correlations despite marked phenotypic differences between Aging and Development. In each interval, ITPKB, PLCD1, PIK3R3, ISYNA1, IMPA2, INPPL1, PI4KB, and AKT1 are in Group 1, PIK3CB, PTEN, PIK3CA, and IMPA1 in Group 2, and SACM1L, PI3KR4, INPP5A, SYNJ1, and PLCB1 in Group 3. Ten of the genes change expression nonlinearly during Development, suggesting involvement in rapidly changing neuronal, glial and myelination events. Correlated transcription for some gene pairs likely is facilitated by colocalization on the same chromosome band. Conclusions Stable coordinated gene transcriptional networks regulate brain phosphoinositide metabolic pathways during human Development and Aging.
Collapse
Affiliation(s)
- Stanley I. Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
- * E-mail:
| | - Christopher T. Primiani
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Chuck T. Chen
- Section on Nutritional Neurosciences, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States of America
| | - Kwangmi Ahn
- Child Psychiatry Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States of America
| | - Veronica H. Ryan
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
39
|
Böhm MRR, Melkonyan H, Thanos S. Life-time expression of the proteins peroxiredoxin, beta-synuclein, PARK7/DJ-1, and stathmin in the primary visual and primary somatosensory cortices in rats. Front Neuroanat 2015; 9:16. [PMID: 25788877 PMCID: PMC4349188 DOI: 10.3389/fnana.2015.00016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/04/2015] [Indexed: 11/16/2022] Open
Abstract
Four distinct proteins are regulated in the aging neuroretina and may be regulated in the cerebral cortex, too: peroxiredoxin, beta-synuclein, PARK[Parkinson disease(autosomal recessive, early onset)]7/DJ-1, and Stathmin. Thus, we performed a comparative analysis of these proteins in the the primary somatosensory cortex (S1) and primary visual cortex (V1) in rats, in order to detect putative common development-, maturation- and age-related changes. The expressions of peroxiredoxin, beta-synuclein, PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1, and Stathmin were compared in the newborn, juvenile, adult, and aged S1 and V1. Western blot (WB), quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and immunohistochemistry (IHC) analyses were employed to determine whether the changes identified by proteomics were verifiable at the cellular and molecular levels. All of the proteins were detected in both of the investigated cortical areas. Changes in the expressions of the four proteins were found throughout the life-time of the rats. Peroxiredoxin expression remained unchanged over life-time. Beta-Synuclein expression was massively increased up to the adult stage of life in both the S1 and V1. PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1 exhibited a massive up-regulation in both the S1 and V1 at all ages. Stathmin expression was massively down regulated after the neonatal period in both the S1 and V1. The detected protein alterations were analogous to their retinal profiles. This study is the first to provide evidence that peroxiredoxin, beta-synuclein, PARK[Parkinson disease (autosomal recessive, early onset)]7/DJ-1, and Stathmin are associated with postnatal maturation and aging in both the S1 and V1 of rats. These changes may indicate their involvement in key functional pathways and may account for the onset or progression of age-related pathologies.
Collapse
Affiliation(s)
- Michael R R Böhm
- Institute of Experimental Ophthalmology and DFG-Center of Excellence Cells in Motion (CiM), area C.4, School of Medicine, Westfalian-Wilhelms-University of Münster Münster, Germany ; Department of Ophthalmology, St. Franziskus Hospital Münster Münster, Germany
| | - Harutyun Melkonyan
- Institute of Experimental Ophthalmology and DFG-Center of Excellence Cells in Motion (CiM), area C.4, School of Medicine, Westfalian-Wilhelms-University of Münster Münster, Germany
| | - Solon Thanos
- Institute of Experimental Ophthalmology and DFG-Center of Excellence Cells in Motion (CiM), area C.4, School of Medicine, Westfalian-Wilhelms-University of Münster Münster, Germany
| |
Collapse
|