1
|
Turtzo LC, Chapagain NY, Peterkin N, Cota MR, Vorn R, Devoto C, O'Keefe J, Emanuel OM, Parikh G, Diaz-Arrastia R, Butman JA, McGavern DB, Chan L, Latour LL. Association of Traumatic Meningeal Enhancement on MRI With Clinical Recovery in Patients With Traumatic Brain Injury. Neurology 2025; 104:e213448. [PMID: 39999394 PMCID: PMC11863783 DOI: 10.1212/wnl.0000000000213448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Traumatic meningeal enhancement (TME) can be observed on MRI of patients with acute traumatic brain injury (TBI) and reflects abnormal contrast extravasation into the meninges. Resolution of TME occurs over time, but TME can persist for weeks, suggesting incomplete meningeal repair. This study's objectives were to describe TME's prevalence, severity, and evolution over time and to investigate TME's association with other imaging findings, blood-based biomarkers commonly associated with TBI, and incomplete recovery. METHODS Patients with suspected TBI presenting within 48 hours of injury to Suburban Hospital or Washington Hospital Center were prospectively enrolled between 2010 and 2019, received MRI, and underwent optional blood collection at baseline and follow-up visits at 1 week, 30 days, 90 days, and 1 year. Independent trained raters blinded to patient details scored for the presence and severity of TME on postcontrast MRI. Neuroimaging findings on CT and MRI, other than TME, were extracted from neuroradiology reports. Plasma biomarker levels (total tau [t-tau]; neurofilament light chain [NfL]; glial fibrillary acidic protein [GFAP]; ubiquitin C-terminal hydrolase-1 [UCH-L1]) were assessed with single-molecule array kits. Incomplete recovery was defined as a Glasgow Outcome Scale-Extended (GOSE) score <7 at 30-90-day follow-up. Factors associated with recovery were assessed through multivariable logistic regression analysis controlled for confounding variables. RESULTS Of 675 patients (male/female/neither 68%/31%/1%; median [interquartile range] age: 45 [28-58] years; Glasgow Coma Scale score 15 [15-15]), 359 (53%) were positive for TME at baseline (16 [6-25] hours after injury). At 30-90-day follow-up, TME remained absent in 117 (37%), resolved in 139 (45%), and persisted in 56 (18%). Acute TME had a high positive predictive value (PPV) for acute TBI-related findings on CT (87.7%) and MRI (86.1%). One-way analysis of covariance demonstrated significant associations between baseline TME and CT for plasma biomarker levels (F(df): t-tau = 19.328 (2); NfL = 20.458 (2); GFAP = 78.662 (2); UCH-LI = 46.680 (2)). Patients with persistent TME were more likely (odds ratio 3.809; 95% CI 1.703-8.519; p = 0.001) to have GOSE score <7. DISCUSSION TME was prevalent at baseline, with high PPV for other neuroimaging findings, but was not associated with recovery. TME's persistence was independently associated with incomplete recovery, suggesting that the meninges' failure to repair may be a mediator of recovery after TBI. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov NCT01132937.
Collapse
Affiliation(s)
- L Christine Turtzo
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
- MTBI2, formerly known as CNRM
| | - Nikita Y Chapagain
- MTBI2, formerly known as CNRM
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | - Nicole Peterkin
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
- MTBI2, formerly known as CNRM
| | - Martin R Cota
- MTBI2, formerly known as CNRM
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | - Rany Vorn
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
- School of Nursing, Johns Hopkins University, Baltimore, MD
| | | | - Jessica O'Keefe
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
- MTBI2, formerly known as CNRM
| | - Olivia M Emanuel
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | - Gunjan Parikh
- R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore
| | | | - John A Butman
- MTBI2, formerly known as CNRM
- Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, MD
| | - Dorian B McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD; and
| | - Leighton Chan
- MTBI2, formerly known as CNRM
- Rehabilitation Medicine Department, Clinical Center, NIH, Bethesda, MD
| | - Lawrence L Latour
- Acute Cerebrovascular Diagnostics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| |
Collapse
|
2
|
Xu D, Zhang C, Bi X, Xu J, Guo S, Li P, Shen Y, Cai J, Zhang N, Tian G, Zhang H, Wang H, Li Q, Jiang H, Wang B, Li X, Li Y, Li K. Mapping enhancer and chromatin accessibility landscapes charts the regulatory network of Alzheimer's disease. Comput Biol Med 2024; 168:107802. [PMID: 38056211 DOI: 10.1016/j.compbiomed.2023.107802] [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: 08/01/2023] [Revised: 10/20/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Enhancers are regulatory elements that target and modulate gene expression and play a role in human health and disease. However, the roles of enhancer regulatory circuit abnormalities driven by epigenetic alterations in Alzheimer's disease (AD) are unclear. METHODS In this study, a multiomic integrative analysis was performed to map enhancer and chromatin accessibility landscapes and identify regulatory network abnormalities in AD. We identified differentially methylated enhancers and constructed regulatory networks across brain regions using AD brain tissue samples. Through the integration of snATAC-seq and snRNA-seq datasets, we mapped enhancers with DNA methylation alterations (DMA) and chromatin accessibility landscapes. Core regulatory triplets that contributed to AD neuropathology in specific cell types were further prioritized. RESULTS We revealed widespread DNA methylation alterations (DMA) in the enhancers of AD patients across different brain regions. In addition, the genome-wide transcription factor (TF) binding profiles showed that enhancers with DMA are pervasively regulated by TFs. The TF-enhancer-gene regulatory network analysis identified core regulatory triplets that are associated with brain and immune cell proportions and play important roles in AD pathogenesis. Enhancer regulatory circuits with DMA exhibited distinct chromatin accessibility patterns, which were further characterized at single-cell resolutions. CONCLUSIONS Our study comprehensively investigated DNA methylation-mediated regulatory circuit abnormalities and provided novel insights into the potential pathogenesis of AD.
Collapse
Affiliation(s)
- Dahua Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Chunrui Zhang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100020, China
| | - Xiaoman Bi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Jiankai Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shengnan Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Peihu Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Yutong Shen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Jiale Cai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Nihui Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Guanghui Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Haifei Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Hong Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Qifu Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Hongyan Jiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Bo Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China.
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China.
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China.
| | - Kongning Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan General Hospital, Hainan Affiliated Hospital, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
3
|
Roberts JA, Varma VR, Candia J, Tanaka T, Ferrucci L, Bennett DA, Thambisetty M. Unbiased proteomics and multivariable regularized regression techniques identify SMOC1, NOG, APCS, and NTN1 in an Alzheimer's disease brain proteomic signature. NPJ AGING 2023; 9:18. [PMID: 37414805 PMCID: PMC10326005 DOI: 10.1038/s41514-023-00112-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
Advancements in omics methodologies have generated a wealth of high-dimensional Alzheimer's disease (AD) datasets, creating significant opportunities and challenges for data interpretation. In this study, we utilized multivariable regularized regression techniques to identify a reduced set of proteins that could discriminate between AD and cognitively normal (CN) brain samples. Utilizing eNetXplorer, an R package that tests the accuracy and significance of a family of elastic net generalized linear models, we identified 4 proteins (SMOC1, NOG, APCS, NTN1) that accurately discriminated between AD (n = 31) and CN (n = 22) middle frontal gyrus (MFG) tissue samples from Religious Orders Study participants with 83 percent accuracy. We then validated this signature in MFG samples from Baltimore Longitudinal Study of Aging participants using leave-one-out logistic regression cross-validation, finding that the signature again accurately discriminated AD (n = 31) and CN (n = 19) participants with a receiver operating characteristic curve area under the curve of 0.863. These proteins were strongly correlated with the burden of neurofibrillary tangle and amyloid pathology in both study cohorts. We additionally tested whether these proteins differed between AD and CN inferior temporal gyrus (ITG) samples and blood serum samples at the time of AD diagnosis in ROS and BLSA, finding that the proteins differed between AD and CN ITG samples but not in blood serum samples. The identified proteins may provide mechanistic insights into the pathophysiology of AD, and the methods utilized in this study may serve as the basis for further work with additional high-dimensional datasets in AD.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Julián Candia
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
4
|
Katzenberger RJ, Ganetzky B, Wassarman DA. Lissencephaly-1 mutations enhance traumatic brain injury outcomes in Drosophila. Genetics 2023; 223:iyad008. [PMID: 36683334 PMCID: PMC9991514 DOI: 10.1093/genetics/iyad008] [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: 11/14/2022] [Revised: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) outcomes vary greatly among individuals, but most of the variation remains unexplained. Using a Drosophila melanogaster TBI model and 178 genetically diverse lines from the Drosophila Genetic Reference Panel (DGRP), we investigated the role that genetic variation plays in determining TBI outcomes. Following injury at 20-27 days old, DGRP lines varied considerably in mortality within 24 h ("early mortality"). Additionally, the disparity in early mortality resulting from injury at 20-27 vs 0-7 days old differed among DGRP lines. These data support a polygenic basis for differences in TBI outcomes, where some gene variants elicit their effects by acting on aging-related processes. Our genome-wide association study of DGRP lines identified associations between single nucleotide polymorphisms in Lissencephaly-1 (Lis-1) and Patronin and early mortality following injury at 20-27 days old. Lis-1 regulates dynein, a microtubule motor required for retrograde transport of many cargoes, and Patronin protects microtubule minus ends against depolymerization. While Patronin mutants did not affect early mortality, Lis-1 compound heterozygotes (Lis-1x/Lis-1y) had increased early mortality following injury at 20-27 or 0-7 days old compared with Lis-1 heterozygotes (Lis-1x/+), and flies that survived 24 h after injury had increased neurodegeneration but an unaltered lifespan, indicating that Lis-1 affects TBI outcomes independently of effects on aging. These data suggest that Lis-1 activity is required in the brain to ameliorate TBI outcomes through effects on axonal transport, microtubule stability, and other microtubule proteins, such as tau, implicated in chronic traumatic encephalopathy, a TBI-associated neurodegenerative disease in humans.
Collapse
Affiliation(s)
- Rebeccah J Katzenberger
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Barry Ganetzky
- Department of Genetics, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Wassarman
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
Amgalan A, Maher AS, Ghosh S, Chui HC, Bogdan P, Irimia A. Brain age estimation reveals older adults' accelerated senescence after traumatic brain injury. GeroScience 2022; 44:2509-2525. [PMID: 35792961 PMCID: PMC9768106 DOI: 10.1007/s11357-022-00597-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/23/2022] [Indexed: 01/06/2023] Open
Abstract
Adults aged 60 and over are most vulnerable to mild traumatic brain injury (mTBI). Nevertheless, the extent to which chronological age (CA) at injury affects TBI-related brain aging is unknown. This study applies Gaussian process regression to T1-weighted magnetic resonance images (MRIs) acquired within [Formula: see text]7 days and again [Formula: see text]6 months after a single mTBI sustained by 133 participants aged 20-83 (CA [Formula: see text] = 42.6 ± 17 years; 51 females). Brain BAs are estimated, modeled, and compared as a function of sex and CA at injury using a statistical model selection procedure. On average, the brains of older adults age by 15.3 ± 6.9 years after mTBI, whereas those of younger adults age only by 1.8 ± 5.6 years, a significant difference (Welch's t32 = - 9.17, p ≃ 9.47 × 10-11). For an adult aged [Formula: see text]30 to [Formula: see text]60, the expected amount of TBI-related brain aging is [Formula: see text]3 years greater than in an individual younger by a decade. For an individual over [Formula: see text]60, the respective amount is [Formula: see text]7 years. Despite no significant sex differences in brain aging (Welch's t108 = 0.78, p > 0.78), the statistical test is underpowered. BAs estimated at acute baseline versus chronic follow-up do not differ significantly (t264 = 0.41, p > 0.66, power = 80%), suggesting negligible TBI-related brain aging during the chronic stage of TBI despite accelerated aging during the acute stage. Our results indicate that a single mTBI sustained after age [Formula: see text]60 involves approximately [Formula: see text]10 years of premature and lasting brain aging, which is MRI detectable as early as [Formula: see text]7 days post-injury.
Collapse
Affiliation(s)
- Anar Amgalan
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Alexander S Maher
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Satyaki Ghosh
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Electronics and Electrical Engineering, Indian Institute of Technology, Guwahati, Assam, India
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paul Bogdan
- Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Corwin D. Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Oestreich LKL, O'Sullivan MJ. Transdiagnostic In Vivo Magnetic Resonance Imaging Markers of Neuroinflammation. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:638-658. [PMID: 35051668 DOI: 10.1016/j.bpsc.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 05/13/2023]
Abstract
Accumulating evidence suggests that inflammation is not limited to archetypal inflammatory diseases such as multiple sclerosis, but instead represents an intrinsic feature of many psychiatric and neurological disorders not typically classified as neuroinflammatory. A growing body of research suggests that neuroinflammation can be observed in early and prodromal stages of these disorders and, under certain circumstances, may lead to tissue damage. Traditional methods to assess neuroinflammation include serum or cerebrospinal fluid markers and positron emission tomography. These methods require invasive procedures or radiation exposure and lack the exquisite spatial resolution of magnetic resonance imaging (MRI). There is, therefore, an increasing interest in noninvasive neuroimaging tools to evaluate neuroinflammation reliably and with high specificity. While MRI does not provide information at a cellular level, it facilitates the characterization of several biophysical tissue properties that are closely linked to neuroinflammatory processes. The purpose of this review is to evaluate the potential of MRI as a noninvasive, accessible, and cost-effective technology to image neuroinflammation across neurological and psychiatric disorders. We provide an overview of current and developing MRI methods used to study different aspects of neuroinflammation and weigh their strengths and shortcomings. Novel MRI contrast agents are increasingly able to target inflammatory processes directly, therefore offering a high degree of specificity, particularly if used in conjunction with multitissue, biophysical diffusion MRI compartment models. The capability of these methods to characterize several aspects of the neuroinflammatory milieu will likely push MRI to the forefront of neuroimaging modalities used to characterize neuroinflammation transdiagnostically.
Collapse
Affiliation(s)
- Lena K L Oestreich
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia.
| | - Michael J O'Sullivan
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; Institute of Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
7
|
Gut bacterial isoamylamine promotes age-related cognitive dysfunction by promoting microglial cell death. Cell Host Microbe 2022; 30:944-960.e8. [PMID: 35654045 DOI: 10.1016/j.chom.2022.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/07/2022] [Accepted: 05/06/2022] [Indexed: 11/21/2022]
Abstract
The intestinal microbiome releases a plethora of small molecules. Here, we show that the Ruminococcaceae metabolite isoamylamine (IAA) is enriched in aged mice and elderly people, whereas Ruminococcaceae phages, belonging to the Myoviridae family, are reduced. Young mice orally administered IAA show cognitive decline, whereas Myoviridae phage administration reduces IAA levels. Mechanistically, IAA promotes apoptosis of microglial cells by recruiting the transcriptional regulator p53 to the S100A8 promoter region. Specifically, IAA recognizes and binds the S100A8 promoter region to facilitate the unwinding of its self-complementary hairpin structure, thereby subsequently enabling p53 to access the S100A8 promoter and enhance S100A8 expression. Thus, our findings provide evidence that small molecules released from the gut microbiome can directly bind genomic DNA and act as transcriptional coregulators by recruiting transcription factors. These findings further unveil a molecular mechanism that connects gut metabolism to gene expression in the brain with implications for disease development.
Collapse
|
8
|
Identification of Regulatory Factors and Prognostic Markers in Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2022; 11:antiox11020303. [PMID: 35204186 PMCID: PMC8868268 DOI: 10.3390/antiox11020303] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/10/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of motor neurons, leading to muscle atrophy, paralysis and even death. Immune disorder, redox imbalance, autophagy disorder, and iron homeostasis disorder have been shown to play critical roles in the pathogenesis of ALS. However, the exact pathogenic genes and the underlying mechanism of ALS remain unclear. The purpose of this study was to screen for pathogenic regulatory genes and prognostic markers in ALS using bioinformatics methods. We used Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, gene set enrichment analysis (GSEA), and expression regulation network analysis to investigate the function of differentially expressed genes in the nerve tissue, lymphoid tissue, and whole blood of patients with ALS. Our results showed that the up-regulated genes were mainly involved in immune regulation and inflammation, and the down-regulated genes were mainly involved in energy metabolism and redox processes. Eleven up-regulated transcription factors (CEBPB, CEBPD, STAT5A, STAT6, RUNX1, REL, SMAD3, GABPB2, FOXO1, PAX6, and FOXJ1) and one down-regulated transcription factor (NOG) in the nerve tissue of patients with ALS likely play important regulatory roles in the pathogenesis of ALS. Based on construction and evaluation of the ALS biomarker screening model, cluster analysis of the identified characteristic genes, univariate Cox proportional hazards regression analysis, and the random survival forest algorithm, we found that MAEA, TPST1, IFNGR2, and ALAS2 may be prognostic markers regarding the survival of ALS patients. High expression of MAEA, TPST1, and IFNGR2 and low expression of ALAS2 in ALS patients may be closely related to short survival of ALS patients. Taken together, our results indicate that immune disorders, inflammation, energy metabolism, and redox imbalance may be the important pathogenic factors of ALS. CEBPB, CEBPD, STAT5A, STAT6, RUNX1, REL, SMAD3, GABPB2, FOXO1, PAX6, FOXJ1, and NOG may be important regulatory factors linked to the pathogenesis of ALS. MAEA, TPST1, IFNGR2, and ALAS2 are potential important ALS prognostic markers. Our findings provide evidence on the pathogenesis of ALS, potential targets for the development of new drugs for ALS, and important markers for predicting ALS prognosis.
Collapse
|
9
|
Moro F, Pischiutta F, Portet A, Needham EJ, Norton EJ, Ferdinand JR, Vegliante G, Sammali E, Pascente R, Caruso E, Micotti E, Tolomeo D, di Marco Barros R, Fraunberger E, Wang KKW, Esser MJ, Menon DK, Clatworthy MR, Zanier ER. Ageing is associated with maladaptive immune response and worse outcome after traumatic brain injury. Brain Commun 2022; 4:fcac036. [PMID: 35350551 PMCID: PMC8947244 DOI: 10.1093/braincomms/fcac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 11/15/2022] Open
Abstract
Traumatic brain injury is increasingly common in older individuals. Older age is one of the strongest predictors for poor prognosis after brain trauma, a phenomenon driven by the presence of extra-cranial comorbidities as well as pre-existent pathologies associated with cognitive impairment and brain volume loss (such as cerebrovascular disease or age-related neurodegeneration). Furthermore, ageing is associated with a dysregulated immune response, which includes attenuated responses to infection and vaccination, and a failure to resolve inflammation leading to chronic inflammatory states. In traumatic brain injury, where the immune response is imperative for the clearance of cellular debris and survey of the injured milieu, an appropriate self-limiting response is vital to promote recovery. Currently, our understanding of age-related factors that contribute to the outcome is limited; but a more complete understanding is essential for the development of tailored therapeutic strategies to mitigate the consequences of traumatic brain injury. Here we show greater functional deficits, white matter abnormalities and worse long-term outcomes in aged compared with young C57BL/6J mice after either moderate or severe traumatic brain injury. These effects are associated with altered systemic, meningeal and brain tissue immune response. Importantly, the impaired acute systemic immune response in the mice was similar to the findings observed in our clinical cohort. Traumatic brain-injured patient cohort over 70 years of age showed lower monocyte and lymphocyte counts compared with those under 45 years. In mice, traumatic brain injury was associated with alterations in peripheral immune subsets, which differed in aged compared with adult mice. There was a significant increase in transcription of immune and inflammatory genes in the meninges post-traumatic brain injury, including monocyte/leucocyte-recruiting chemokines. Immune cells were recruited to the region of the dural injury, with a significantly higher number of CD11b+ myeloid cells in aged compared with the adult mice. In brain tissue, when compared with the young adult mice, we observed a more pronounced and widespread reactive astrogliosis 1 month after trauma in aged mice, sustained by an early and persistent induction of proinflammatory astrocytic state. These findings provide important insights regarding age-related exacerbation of neurological damage after brain trauma.
Collapse
Affiliation(s)
- Federico Moro
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Francesca Pischiutta
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Anaïs Portet
- Molecular Immunity Unit, Department of Medicine, Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, UK
| | - Edward J. Needham
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QH, UK
| | - Emma J. Norton
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QH, UK
| | - John R. Ferdinand
- Molecular Immunity Unit, Department of Medicine, Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, UK
| | - Gloria Vegliante
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Eliana Sammali
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Rosaria Pascente
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Enrico Caruso
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Edoardo Micotti
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Daniele Tolomeo
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Rafael di Marco Barros
- Molecular Immunity Unit, Department of Medicine, Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, UK
| | - Erik Fraunberger
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
- Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Kevin K. W. Wang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, FL, USA
| | - Michael J. Esser
- Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - David K. Menon
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QH, UK
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, Laboratory of Molecular Biology, University of Cambridge, Cambridge CB2 0QH, UK
| | - Elisa R. Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
- Correspondence to: Elisa R. Zanier Laboratory of Acute Brain Injury and Therapeutic Strategies Department of Neuroscience Istituto di Ricerche Farmacologiche Mario Negri IRCCS 20156 Milan, Italy E-mail:
| |
Collapse
|
10
|
Jackson TC, Kochanek PM. RNA Binding Motif 5 (RBM5) in the CNS-Moving Beyond Cancer to Harness RNA Splicing to Mitigate the Consequences of Brain Injury. Front Mol Neurosci 2020; 13:126. [PMID: 32765218 PMCID: PMC7381114 DOI: 10.3389/fnmol.2020.00126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Gene splicing modulates the potency of cell death effectors, alters neuropathological disease processes, influences neuronal recovery, but may also direct distinct mechanisms of secondary brain injury. Therapeutic targeting of RNA splicing is a promising avenue for next-generation CNS treatments. RNA-binding proteins (RBPs) regulate a variety of RNA species and are prime candidates in the hunt for druggable targets to manipulate and tailor gene-splicing responses in the brain. RBPs preferentially recognize unique consensus sequences in targeted mRNAs. Also, RBPs often contain multiple RNA-binding domains (RBDs)—each having a unique consensus sequence—suggesting the possibility that drugs could be developed to block individual functional domains, increasing the precision of RBP-targeting therapies. Empirical characterization of most RBPs is lacking and represents a major barrier to advance this emerging therapeutic area. There is a paucity of data on the role of RBPs in the brain including, identification of their unique mRNA targets, defining how CNS insults affect their levels and elucidating which RBPs (and individual domains within) to target to improve neurological outcomes. This review focuses on the state-of-the-art of the RBP tumor suppressor RNA binding motif 5 (RBM5) in the CNS. We discuss its potent pro-death roles in cancer, which motivated our interest to study it in the brain. We review recent studies showing that RBM5 levels are increased after CNS trauma and that it promotes neuronal death in vitro. Finally, we conclude with recent reports on the first set of RBM5 regulated genes identified in the intact brain, and discuss how those findings provide new clues germane to its potential function(s) in the CNS, and pose new questions on its therapeutic utility to mitigate CNS injury.
Collapse
Affiliation(s)
- Travis C Jackson
- Morsani College of Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, United States.,Morsani College of Medicine, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Identification of Methylated Gene Biomarkers in Patients with Alzheimer's Disease Based on Machine Learning. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8348147. [PMID: 32309439 PMCID: PMC7139879 DOI: 10.1155/2020/8348147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/12/2020] [Accepted: 03/03/2020] [Indexed: 11/17/2022]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder and characterized by the cognitive impairments. It is essential to identify potential gene biomarkers for AD pathology. Methods DNA methylation expression data of patients with AD were downloaded from the Gene Expression Omnibus (GEO) database. Differentially methylated sites were identified. The functional annotation analysis of corresponding genes in the differentially methylated sites was performed. The optimal diagnostic gene biomarkers for AD were identified by using random forest feature selection procedure. In addition, receiver operating characteristic (ROC) diagnostic analysis of differentially methylated genes was performed. Results A total of 10 differentially methylated sites including 5 hypermethylated sites and 5 hypomethylated sites were identified in AD. There were a total of 8 genes including thioredoxin interacting protein (TXNIP), noggin (NOG), regulator of microtubule dynamics 2 (FAM82A1), myoneurin (MYNN), ankyrin repeat domain 34B (ANKRD34B), STAM-binding protein like 1, ALMalpha (STAMBPL1), cyclin-dependent kinase inhibitor 1C (CDKN1C), and coronin 2B (CORO2B) that correspond to 10 differentially methylated sites. The cell cycle (FDR = 0.0284087) and TGF-beta signaling pathway (FDR = 0.0380372) were the only two significantly enriched pathways of these genes. MYNN was selected as optimal diagnostic biomarker with great diagnostic value. The random forests model could effectively predict AD. Conclusion Our study suggested that MYNN could be served as optimal diagnostic biomarker of AD. Cell cycle and TGF-beta signaling pathway may be associated with AD.
Collapse
|
12
|
Edwards KA, Motamedi V, Osier ND, Kim HS, Yun S, Cho YE, Lai C, Dell KC, Carr W, Walker P, Ahlers S, LoPresti M, Yarnell A, Tschiffley A, Gill JM. A Moderate Blast Exposure Results in Dysregulated Gene Network Activity Related to Cell Death, Survival, Structure, and Metabolism. Front Neurol 2020; 11:91. [PMID: 32174881 PMCID: PMC7054450 DOI: 10.3389/fneur.2020.00091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Blast exposure is common in military personnel during training and combat operations, yet biological mechanisms related to cell survival and function that coordinate recovery remain poorly understood. This study explored how moderate blast exposure influences gene expression; specifically, gene-network changes following moderate blast exposure. On day 1 (baseline) of a 10-day military training program, blood samples were drawn, and health and demographic information collected. Helmets equipped with bilateral sensors worn throughout training measured overpressure in pounds per square inch (psi). On day 7, some participants experienced moderate blast exposure (peak pressure ≥5 psi). On day 10, 3 days post-exposure, blood was collected and compared to baseline with RNA-sequencing to establish gene expression changes. Based on dysregulation data from RNA-sequencing, followed by top gene networks identified with Ingenuity Pathway Analysis, a subset of genes was validated (NanoString). Five gene networks were dysregulated; specifically, two highly significant networks: (1) Cell Death and Survival (score: 42), including 70 genes, with 50 downregulated and (2) Cell Structure, Function, and Metabolism (score: 41), including 69 genes, with 41 downregulated. Genes related to ubiquitination, including neuronal development and repair: UPF1, RNA Helicase and ATPase (UPF1) was upregulated while UPF3 Regulator of Nonsense Transcripts Homolog B (UPF3B) was downregulated. Genes related to inflammation were upregulated, including AKT serine/threonine kinase 1 (AKT1), a gene coordinating cellular recovery following TBIs. Moderate blast exposure induced significant gene expression changes including gene networks involved in (1) cell death and survival and (2) cellular development and function. The present findings may have implications for understanding blast exposure pathology and subsequent recovery efforts.
Collapse
Affiliation(s)
- Katie A Edwards
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Vida Motamedi
- Wake Forest School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Nicole D Osier
- School of Nursing, University of Texas at Austin, Austin, TX, United States.,Department of Neurology, University of Texas, Austin, TX, United States
| | - Hyung-Suk Kim
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Sijung Yun
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Young-Eun Cho
- College of Nursing, University of Iowa, Iowa City, IA, United States
| | - Chen Lai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Kristine C Dell
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Walter Carr
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Peter Walker
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Stephen Ahlers
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Matthew LoPresti
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Angela Yarnell
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Anna Tschiffley
- Naval Medical Research Center, Silver Spring, MD, United States
| | - Jessica M Gill
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,CNRM Co-Director Biomarkers Core, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
Zamani A, Powell KL, May A, Semple BD. Validation of reference genes for gene expression analysis following experimental traumatic brain injury in a pediatric mouse model. Brain Res Bull 2020; 156:43-49. [PMID: 31904409 DOI: 10.1016/j.brainresbull.2019.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/02/2019] [Accepted: 12/30/2019] [Indexed: 01/01/2023]
Abstract
Quantitative polymerase chain reaction (qPCR) is the gold standard method in targeted analysis of messenger RNA (mRNA) levels in a tissue. To minimize methodological errors, a reference gene (or a combination of reference genes) is routinely used for normalization to account for technical variables such as RNA quality and sample size. While presumed to have stable expression, reference genes in the brain can change during normal development, as well as in response to injury, such as traumatic brain injury (TBI). This study is the first to evaluate the stability of reference genes in a controlled cortical impact (CCI) model in the pediatric mouse brain, using two methods of qPCR normalization for optimal reference gene selection. Three week old mice were subjected to unilateral CCI at two severity of injuries (mild or severe), compared to sham controls. At 1 and 8 weeks post-injury, the ipsilateral hemisphere was analyzed to determine reference gene stability. Five commonly-used reference genes were compared: tyrosine 3 monooxygenase/tryptophan 5 monooxygenase activation protein zeta (Ywhaz), cyclophilin A (Ppia), hypoxanthine phosphoribosyl transferase (Hprt), glyceraldehyde-3-phosphate dehydrogenase (Gapdh) and β-actin (Actb). Ppia and Hprt were chosen as the most stable combination of genes using GeNORM software analysis. These results highlight the instability of several commonly used reference genes after TBI, and provide a selection of validated genes for future gene expression analyses in the injured pediatric mouse brain.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia.
| | - Kim L Powell
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Ashleigh May
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia; Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, 3052, Australia
| |
Collapse
|
14
|
Sakharkar MK, Kashmir Singh SK, Rajamanickam K, Mohamed Essa M, Yang J, Chidambaram SB. A systems biology approach towards the identification of candidate therapeutic genes and potential biomarkers for Parkinson's disease. PLoS One 2019; 14:e0220995. [PMID: 31487305 PMCID: PMC6728017 DOI: 10.1371/journal.pone.0220995] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is an irreversible and incurable multigenic neurodegenerative disorder. It involves progressive loss of mid brain dopaminergic neurons in the substantia nigra pars compacta (SN). We compared brain gene expression profiles with those from the peripheral blood cells of a separate sample of PD patients to identify disease-associated genes. Here, we demonstrate the use of gene expression profiling of brain and blood for detecting valid targets and identifying early PD biomarkers. Implementing this systematic approach, we discovered putative PD risk genes in brain, delineated biological processes and molecular functions that may be particularly disrupted in PD and also identified several putative PD biomarkers in blood. 20 of the differentially expressed genes in SN were also found to be differentially expressed in the blood. Further application of this methodology to other brain regions and neurological disorders should facilitate the discovery of highly reliable and reproducible candidate risk genes and biomarkers for PD. The identification of valid peripheral biomarkers for PD may ultimately facilitate early identification, intervention, and prevention efforts as well.
Collapse
Affiliation(s)
- Meena Kishore Sakharkar
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
- * E-mail: (MKS); (SBC)
| | | | - Karthic Rajamanickam
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | | | - Jian Yang
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSSAHER, Karnataka, India
- * E-mail: (MKS); (SBC)
| |
Collapse
|
15
|
Cheng X, Ferino E, Hull H, Jickling GC, Ander BP, Stamova B, Sharp FR. Smoking affects gene expression in blood of patients with ischemic stroke. Ann Clin Transl Neurol 2019; 6:1748-1756. [PMID: 31436916 PMCID: PMC6764500 DOI: 10.1002/acn3.50876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/27/2019] [Accepted: 07/27/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Though cigarette smoking (CS) is a well-known risk factor for ischemic stroke (IS), there is no data on how CS affects the blood transcriptome in IS patients. METHODS We recruited IS-current smokers (IS-SM), IS-never smokers (IS-NSM), control-smokers (C-SM), and control-never smokers (C-NSM). mRNA expression was assessed on HTA-2.0 microarrays and unique as well as commonly expressed genes identified for IS-SM versus IS-NSM and C-SM versus C-NSM. RESULTS One hundred and fifty-eight genes were differentially expressed in IS-SM versus IS-NSM; 100 genes were differentially expressed in C-SM versus C-NSM; and 10 genes were common to both IS-SM and C-SM (P < 0.01; |fold change| ≥ 1.2). Functional pathway analysis showed the 158 IS-SM-regulated genes were associated with T-cell receptor, cytokine-cytokine receptor, chemokine, adipocytokine, tight junction, Jak-STAT, ubiquitin-mediated proteolysis, and adherens junction signaling. IS-SM showed more altered genes and functional networks than C-SM. INTERPRETATION We propose some of the 10 genes that are elevated in both IS-SM and C-SM (GRP15, LRRN3, CLDND1, ICOS, GCNT4, VPS13A, DAP3, SNORA54, HIST1H1D, and SCARNA6) might contribute to increased risk of stroke in current smokers, and some genes expressed by blood leukocytes and platelets after stroke in smokers might contribute to worse stroke outcomes that occur in smokers.
Collapse
Affiliation(s)
- Xiyuan Cheng
- Department of Neurology, University of California at Davis, Sacramento, California.,Toxicology and Pharmacology Graduate Program, University of California at Davis, Davis, California
| | - Eva Ferino
- Department of Neurology, University of California at Davis, Sacramento, California
| | - Heather Hull
- Department of Neurology, University of California at Davis, Sacramento, California
| | - Glen C Jickling
- Department of Neurology, University of California at Davis, Sacramento, California.,Department of Neurology, University of Alberta, Edmonton, California
| | - Bradley P Ander
- Department of Neurology, University of California at Davis, Sacramento, California
| | - Boryana Stamova
- Department of Neurology, University of California at Davis, Sacramento, California
| | - Frank R Sharp
- Department of Neurology, University of California at Davis, Sacramento, California.,Toxicology and Pharmacology Graduate Program, University of California at Davis, Davis, California
| |
Collapse
|
16
|
Sparkman NL, Buchanan JB, Dos Santos NL, Johnson RW, Burton MD. Aging sensitizes male mice to cognitive dysfunction induced by central HIV-1 gp120. Exp Gerontol 2019; 126:110694. [PMID: 31437586 DOI: 10.1016/j.exger.2019.110694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/04/2019] [Accepted: 08/17/2019] [Indexed: 10/26/2022]
Abstract
Although highly active antiretroviral therapy has led to improved prognosis and alleviation of some HIV-related disease complications, it has not provided complete protection against HIV-associated dementia. As the population of persons living with HIV grows older and aged persons represent a significant number of new infections, it is important to understand how HIV may affect the aged brain. In the current study, both adult and aged mice were treated with HIV gp120 and trained in a reference memory version of the water maze. Analysis of probe data revealed that aged animals treated with gp120 demonstrated profound decrements in water maze performance compared to gp120 treated young animals and saline treated aged or young animals. Additionally, we examined the neuroinflammatory responses in the aged and adult brain 4 h after treatment with gp120. Pro-inflammatory cytokines associated with neuroinflammation are known to be antagonistic to learning and memory processes and aged and adult animals treated with gp120 demonstrated similar increases in IL-1β and IL-6 in the hippocampus and cortex. Additionally, gp120 treatment was associated with an increase in MHCII gene expression, a marker of microglial activation, in the hippocampus. Although, the aged brain demonstrated a similar inflammatory profile at the time point measured, aged animals were more sensitive to cognitive dysfunction related to gp120 treatment. This finding supports the theory that aging may be a significant risk factor in the development of HIV-associated dementia.
Collapse
Affiliation(s)
- Nathan L Sparkman
- Department of Psychology, Stephen F. Austin, PO Box 13046, SFA Station, Nacogdoches, TX 75962, United States of America.
| | - Jessica B Buchanan
- Laboratory of Integrative Immunology and Behavior, Department of Animal Sciences, University of Illinois Urbana-Champaign,1207 W. Gregory Drive, Urbana, IL 61801, United States of America
| | - Natalia L Dos Santos
- Neuroimmunology and Behavior Laboratory, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, BSB 10.536, 800 W Campbell Rd., Richardson, TX 75080, United States of America
| | - Rodney W Johnson
- Laboratory of Integrative Immunology and Behavior, Department of Animal Sciences, University of Illinois Urbana-Champaign,1207 W. Gregory Drive, Urbana, IL 61801, United States of America
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, BSB 10.536, 800 W Campbell Rd., Richardson, TX 75080, United States of America.
| |
Collapse
|
17
|
de Freitas Cardoso MG, Faleiro RM, de Paula JJ, Kummer A, Caramelli P, Teixeira AL, de Souza LC, Miranda AS. Cognitive Impairment Following Acute Mild Traumatic Brain Injury. Front Neurol 2019; 10:198. [PMID: 30906278 PMCID: PMC6418036 DOI: 10.3389/fneur.2019.00198] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/15/2019] [Indexed: 01/28/2023] Open
Abstract
Patients with mild traumatic brain injury (mTBI) may present cognitive deficits within the first 24 h after trauma, herein called "acute phase," which in turn may lead to long-term functional impairment and decrease in quality of life. Few studies investigated cognition in mTBI patients during the acute phase. The objectives of this study were to investigate the cognitive profile of patients with mTBI during the acute phase, compared to controls and normative data, and whether loss of consciousness (LOC), previous TBI and level of education influence cognition at this stage. Fifty-three patients with mTBI (aged 19-64 years) and 28 healthy controls participated in the study. All patients were evaluated at bedside within 24 h post-injury. Demographic and clinical data were registered. Cognitive function was assessed with the Mini-mental state examination (MMSE), the Frontal Assessment Battery (FAB), Digit Span (working memory), and the Visual Memory Test/Brief Cognitive Battery (for episodic memory). The clinical sample was composed mainly by men (58.5%). The mean age was 39 years-old and 64.3% of the patients had more than 8 years of education. The most common causes of mTBI were fall from own height (28.3%), aggression (24.5%), and fall from variable heights (24.5%). Compared to controls, mTBI patients exhibited significantly worse performance on MMSE, FAB, naming, incidental memory, immediate memory, learning, and delayed recall. Compared to normative data, 26.4% of patients had reduced global cognition as measured by the MMSE. Episodic memory impairment (13.2%) was more frequent than executive dysfunction (9.4%). No significant differences were found in cognitive performance when comparing patients with or without LOC or those with or without history of previous TBI. Patients with lower educational level had higher rates of cognitive impairment (VMT naming-28.6 vs. 4.2%; VMT immediate memory-32 vs. 4.2%; VMT learning-39.3 vs. 4.2%, all p < 0.05). In sum, we found significant cognitive impairment in the acute phase of mTBI, which was not associated with LOC or history of TBI, but appeared more frequently in patients with lower educational level.
Collapse
Affiliation(s)
- Maíra Glória de Freitas Cardoso
- Neuroscience Program, Laboratório Interdisciplinar em Investigação Médica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Moreira Faleiro
- Faculdade de Ciências Médicas de Minas Gerais, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Brazil
| | | | - Arthur Kummer
- Laboratório Interdisciplinar em Investigação Médica, Eli Lilly and Company do Brasil, São Paulo, Brazil
| | - Paulo Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Lúcio Teixeira
- Santa Casa BH Ensino e Pesquisa, Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Leonardo Cruz de Souza
- Laboratório Interdisciplinar em Investigação Médica, Departamento de Clínica Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline Silva Miranda
- Laboratório Interdisciplinar em Investigação Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
18
|
Irimia A, Van Horn JD, Vespa PM. Cerebral microhemorrhages due to traumatic brain injury and their effects on the aging human brain. Neurobiol Aging 2018; 66:158-164. [PMID: 29579686 PMCID: PMC5924627 DOI: 10.1016/j.neurobiolaging.2018.02.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/24/2018] [Accepted: 02/27/2018] [Indexed: 01/08/2023]
Abstract
Although cerebral microbleeds (CMBs) are frequently associated with traumatic brain injury (TBI), their effects on clinical outcome after TBI remain controversial and poorly understood, particularly in older adults. Here we (1) highlight major challenges and opportunities associated with studying the effects of TBI-mediated CMBs; (2) review the evidence on their potential effects on cognitive and neural outcome as a function of age at injury; and (3) suggest priorities for future research on understanding the clinical implications of CMBs. Although TBI-mediated CMBs are likely distinct from those due to cerebral amyloid angiopathy or other neurodegenerative diseases, the effects of these 2 CMB types on brain function may share common features. Furthermore, in older TBI victims, the incidence of TBI-mediated CMBs may approximate that of cerebral amyloid angiopathy-related CMBs, and thus warrants detailed study. Because the alterations effected by CMBs on brain structure and function are both unique and age-dependent, it seems likely that novel, age-tailored therapeutic approaches are necessary for the adequate clinical interpretation and treatment of these ubiquitous and underappreciated TBI sequelae.
Collapse
Affiliation(s)
- Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles CA, USA.
| | - John D Van Horn
- USC Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Paul M Vespa
- Departments of Neurosurgery and Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
19
|
Nichols JN, Hagan KL, Floyd CL. Evaluation of Touchscreen Chambers To Assess Cognition in Adult Mice: Effect of Training and Mild Traumatic Brain Injury. J Neurotrauma 2018; 34:2481-2494. [PMID: 28558476 DOI: 10.1089/neu.2017.4998] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cognitive impairments are often experienced after a mild traumatic brain injury (mTBI). In the clinical arena, neuropsychological assessments are used frequently to detect cognitive deficits. Animal models of mTBI, however, rely on an assortment of behavioral tasks to assess cognitive outcome. Computer-based touchscreen systems have been developed for rodents and are hypothesized to offer a translational approach to evaluate cognitive function because of the similarities of tasks performed in rodents to those implemented in humans. While these touchscreen systems have been used in pre-clinical models of neurodegenerative diseases and psychiatric disorders, their use in assessing cognitive impairment after mTBI has not been investigated. We hypothesized that mTBI would result in impaired cognitive performance on touchscreen tasks, particularly those with hippocampal-based learning components, including the paired associate learning (PAL) task and the location discrimination (LD) task. Adult male, C57BL/6 mice received a single impact-acceleration mTBI. We found that training mice before injury to perform to criteria is arduous and that performance is sensitive to many environmental variables. Despite extensive optimization and training, mice failed to perform better than chance in the PAL paradigm. Alternatively, mice demonstrated some capacity to learn in the LD paradigm, but only with the easier stages of the task. The mTBI did not affect performance in the LD paradigm, however. Thus, we concluded that under the conditions presented here, the PAL and LD touchscreen tasks are not robust outcome measures for the evaluation of cognitive performance in C57BL/6 mice after a single impact-acceleration mTBI.
Collapse
Affiliation(s)
- Jessica N Nichols
- 1 Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Kenton L Hagan
- 2 Department of Physical Medicine and Rehabilitation, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Candace L Floyd
- 1 Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
20
|
Barcelona de Mendoza V, Huang Y, Crusto CA, Sun YV, Taylor JY. Perceived Racial Discrimination and DNA Methylation Among African American Women in the InterGEN Study. Biol Res Nurs 2018; 20:145-152. [PMID: 29258399 PMCID: PMC5741522 DOI: 10.1177/1099800417748759] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Experiences of racial discrimination have been associated with poor health outcomes. Little is known, however, about how perceived racial discrimination influences DNA methylation (DNAm) among African Americans (AAs). We examined the association of experiences of discrimination with DNAm among AA women in the Intergenerational Impact of Genetic and Psychological Factors on Blood Pressure (InterGEN) study. METHODS The InterGEN study examines the effects of genetic and psychological factors on blood pressure among AA women and their children. Measures include the Major Life Discrimination (MLD) and the Race-Related Events (RES) scales. In the present analysis, we examined discrimination and DNAm at baseline in the InterGEN study. The 850K EPIC Illumina BeadChip was used for evaluating DNAm in this epigenome-wide association study (EWAS). RESULTS One hundred and fifty-two women contributed data for the RES-EWAS analysis and 147 for the MLD-EWAS analysis. Most were 30-39 years old, nonsmokers, had some college education, and had incomes CONCLUSION We observed significant epigenetic associations between disease-associated genes (e.g., schizophrenia, bipolar disorder, and asthma) and perceived discrimination as measured by the MLD Scale. Future health disparities research should include epigenetics in high-risk populations to elucidate functional consequences induced by the psychosocial environment.
Collapse
Affiliation(s)
| | - Yunfeng Huang
- 2 Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Cindy A Crusto
- 3 Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Yan V Sun
- 2 Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | | |
Collapse
|