51
|
Zhang X, Ma L, Liang D, Song B, Chen J, Huang Y, Xu L, Zhao P, Wu W, Zhang N, Xue R. Neurofilament Light Protein Predicts Disease Progression in Idiopathic REM Sleep Behavior Disorder. JOURNAL OF PARKINSON'S DISEASE 2023:JPD223519. [PMID: 37182898 DOI: 10.3233/jpd-223519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Idiopathic rapid eye movement sleep behavior disorder (iRBD) is increasingly recognized as a manifestation preceding the α-synucleinopathies like Parkinson's disease (PD). Neurofilament light chain (NfL) have been reported to be higher in synucleinopathies as a sign of neurodegeneration. OBJECTIVE To evaluate whether plasma NfL is valuable in reflecting cognitive and motor status in iRBD and PD with a premorbid history of RBD (PDRBD), and predicting disease progression in iRBD. METHODS Thirty-one patients with iRBD, 30 with PDRBD, and 18 healthy controls were included in the cross-sectional and prospective study. Another cohort from the Parkinson's Progression Markers Initiative (PPMI) dataset was enrolled for verification analysis. All patients received evaluations of cognitive, motor, and autonomic function by a battery of clinical tests at baseline and follow-up. Blood NfL was measured by the Quanterix Simoa HD-1. RESULTS In our cohort, 26 patients with iRBD completed the follow-up evaluations, among whom eight (30.8%) patients displayed phenoconversion. Baseline plasma NfL cutoff value of 22.93 pg/mL performed best in distinguishing the iRBD converters from non-converters (sensitivity: 75.0%, specificity: 83.3%, area under the curve: 0.84). Cognitive and motor function were significantly correlated with NfL levels in PDRBD (correlation coefficients: -0.379, 0.399; respectively). Higher baseline NfL levels in iRBD were significantly associated with higher risks for cognitive, motor, autonomic function progression, and phenoconversion at follow-up (hazard ratios: 1.069, 1.065, 1.170, 1.065; respectively). The findings were supported by the PPMI dataset. CONCLUSION Plasma NfL is valuable in reflecting disease severity of PDRBD and predicting disease progression and phenoconversion in iRBD.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Li Ma
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Danqi Liang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingxin Song
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingshan Chen
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaqin Huang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Wu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Xue
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
52
|
Ciardullo S, Muraca E, Bianconi E, Ronchetti C, Cannistraci R, Rossi L, Perra S, Zerbini F, Perseghin G. Serum neurofilament light chain levels are associated with all-cause mortality in the general US population. J Neurol 2023:10.1007/s00415-023-11739-6. [PMID: 37085649 DOI: 10.1007/s00415-023-11739-6] [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: 03/13/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
INTRODUCTION Serum neurofilament light chain (sNfL) levels are biomarkers of neuro-axonal injury in multiple neurological diseases. Little is known on their potential role as prognostic markers in people without known neurological conditions. OBJECTIVE The aim of this study is to evaluate the association between sNfL levels and all-cause mortality in a general population setting. METHODS sNfL levels were measured in 2071 people aged 25-75 years from the general US population that participated in the 2013-2014 cycles of the National Health and Nutrition Examination Survey (NHANES). Cognitive function was evaluated in a subset of participants aged 60-75 years using the Consortium to Establish a Registry for Alzheimer's Disease-Word Learning test, the Animal Fluency test and the Digit Symbol Substitution test. We applied Cox proportional hazard models adjusted for several potential confounders to evaluate the association between sNfL and all-cause mortality through December 2019 by linking NHANES data with data from the National Death Index. RESULTS In a cross-sectional analysis, higher sNfL levels were associated with worse performance in all three cognitive function tests. Over a median follow-up of 6.1 years, 85 participants died. In a multivariable model adjusted for age, sex, race-ethnicity, diabetes, chronic kidney disease, harmful alcohol consumption, cigarette smoke and prevalent cardiovascular disease, higher sNfL levels were significantly and positively associated with all-cause mortality (HR per unit increase in log-transformed sNfL: 2.46, 95% CI 1.77-3.43, p < 0.001). Results were robust when analyses were stratified according to age, sex, body mass index and kidney function. CONCLUSION We found a positive association between sNfL levels and mortality in the general US population. Further studies are needed to understand the biological mechanisms underlying this association.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy.
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy.
| | - Emanuele Muraca
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Eleonora Bianconi
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Celeste Ronchetti
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Rosa Cannistraci
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Laura Rossi
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Silvia Perra
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Francesca Zerbini
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900, Monza, MB, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| |
Collapse
|
53
|
Youssef P, Hughes L, Kim WS, Halliday GM, Lewis SJG, Cooper A, Dzamko N. Evaluation of plasma levels of NFL, GFAP, UCHL1 and tau as Parkinson's disease biomarkers using multiplexed single molecule counting. Sci Rep 2023; 13:5217. [PMID: 36997567 PMCID: PMC10063670 DOI: 10.1038/s41598-023-32480-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 03/28/2023] [Indexed: 04/01/2023] Open
Abstract
Objective biomarkers for Parkinson's Disease (PD) could aid early and specific diagnosis, effective monitoring of disease progression, and improved design and interpretation of clinical trials. Although alpha-synuclein remains a biomarker candidate of interest, the multifactorial and heterogenous nature of PD highlights the need for a PD biomarker panel. Ideal biomarker candidates include markers that are detectable in easily accessible samples, (ideally blood) and that reflect the underlying pathological process of PD. In the present study, we explored the diagnostic and prognostic PD biomarker potential of the SIMOA neurology 4-plex-A biomarker panel, which included neurofilament light (NFL), glial fibrillary acid protein (GFAP), tau and ubiquitin C-terminal hydrolase L1 (UCHL-1). We initially performed a serum vs plasma comparative study to determine the most suitable blood-based matrix for the measurement of these proteins in a multiplexed assay. The levels of NFL and GFAP in plasma and serum were highly correlated (Spearman rho-0.923, p < 0.0001 and rho = 0.825, p < 0.001 respectively). In contrast, the levels of tau were significantly higher in plasma compared to serum samples (p < 0.0001) with no correlation between sample type (Spearman p > 0.05). The neurology 4-plex-A panel, along with plasma alpha-synuclein was then assessed in a cross-sectional cohort of 29 PD patients and 30 controls. Plasma NFL levels positively correlated with both GFAP and alpha-synuclein levels (rho = 0.721, p < 0.0001 and rho = 0.390, p < 0.05 respectively). As diagnostic biomarkers, the control and PD groups did not differ in their mean NFL, GFAP, tau or UCHL-1 plasma levels (t test p > 0.05). As disease state biomarkers, motor severity (MDS-UPDRS III) correlated with increased NFL (rho = 0.646, p < 0.0001), GFAP (rho = 0.450, p < 0.05) and alpha-synuclein levels (rho = 0.406, p < 0.05), while motor stage (Hoehn and Yahr) correlated with increased NFL (rho = 0.455, p < 0.05) and GFAP (rho = 0.549, p < 0.01) but not alpha-synuclein levels (p > 0.05). In conclusion, plasma was determined to be most suitable blood-based matrix for multiplexing the neurology 4-plex-A panel. Given their correlation with motor features of PD, NFL and GFAP appear to be promising disease state biomarker candidates and further longitudinal validation of these two proteins as blood-based biomarkers for PD progression is warranted.
Collapse
Affiliation(s)
- Priscilla Youssef
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Laura Hughes
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Woojin S Kim
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Glenda M Halliday
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Simon J G Lewis
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Antony Cooper
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, UNSW-Sydney, Darlinghurst, NSW, 2010, Australia
| | - Nicolas Dzamko
- Faculty of Medicine and Health and the Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
54
|
Gugger JJ, Sinha N, Huang Y, Walter AE, Lynch C, Kalyani P, Smyk N, Sandsmark D, Diaz-Arrastia R, Davis KA. Structural brain network deviations predict recovery after traumatic brain injury. Neuroimage Clin 2023; 38:103392. [PMID: 37018913 PMCID: PMC10122019 DOI: 10.1016/j.nicl.2023.103392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/10/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVE Traumatic brain injury results in diffuse axonal injury and the ensuing maladaptive alterations in network function are associated with incomplete recovery and persistent disability. Despite the importance of axonal injury as an endophenotype in TBI, there is no biomarker that can measure the aggregate and region-specific burden of axonal injury. Normative modeling is an emerging quantitative case-control technique that can capture region-specific and aggregate deviations in brain networks at the individual patient level. Our objective was to apply normative modeling in TBI to study deviations in brain networks after primarily complicated mild TBI and study its relationship with other validated measures of injury severity, burden of post-TBI symptoms, and functional impairment. METHOD We analyzed 70 T1-weighted and diffusion-weighted MRIs longitudinally collected from 35 individuals with primarily complicated mild TBI during the subacute and chronic post-injury periods. Each individual underwent longitudinal blood sampling to characterize blood protein biomarkers of axonal and glial injury and assessment of post-injury recovery in the subacute and chronic periods. By comparing the MRI data of individual TBI participants with 35 uninjured controls, we estimated the longitudinal change in structural brain network deviations. We compared network deviation with independent measures of acute intracranial injury estimated from head CT and blood protein biomarkers. Using elastic net regression models, we identified brain regions in which deviations present in the subacute period predict chronic post-TBI symptoms and functional status. RESULTS Post-injury structural network deviation was significantly higher than controls in both subacute and chronic periods, associated with an acute CT lesion and subacute blood levels of glial fibrillary acid protein (r = 0.5, p = 0.008) and neurofilament light (r = 0.41, p = 0.02). Longitudinal change in network deviation associated with change in functional outcome status (r = -0.51, p = 0.003) and post-concussive symptoms (BSI: r = 0.46, p = 0.03; RPQ: r = 0.46, p = 0.02). The brain regions where the node deviation index measured in the subacute period predicted chronic TBI symptoms and functional status corresponded to areas known to be susceptible to neurotrauma. CONCLUSION Normative modeling can capture structural network deviations, which may be useful in estimating the aggregate and region-specific burden of network changes induced by TAI. If validated in larger studies, structural network deviation scores could be useful for enrichment of clinical trials of targeted TAI-directed therapies.
Collapse
Affiliation(s)
- James J Gugger
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Neuroengineering & Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Nishant Sinha
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Neuroengineering & Therapeutics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yiming Huang
- Interdisciplinary Computing and Complex BioSystems, School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexa E Walter
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cillian Lynch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyanka Kalyani
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan Smyk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle Sandsmark
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn A Davis
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Neuroengineering & Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
55
|
Höhn L, Hußler W, Richter A, Smalla KH, Birkl-Toeglhofer AM, Birkl C, Vielhaber S, Leber SL, Gundelfinger ED, Haybaeck J, Schreiber S, Seidenbecher CI. Extracellular Matrix Changes in Subcellular Brain Fractions and Cerebrospinal Fluid of Alzheimer’s Disease Patients. Int J Mol Sci 2023; 24:ijms24065532. [PMID: 36982604 PMCID: PMC10058969 DOI: 10.3390/ijms24065532] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
The brain’s extracellular matrix (ECM) is assumed to undergo rearrangements in Alzheimer’s disease (AD). Here, we investigated changes of key components of the hyaluronan-based ECM in independent samples of post-mortem brains (N = 19), cerebrospinal fluids (CSF; N = 70), and RNAseq data (N = 107; from The Aging, Dementia and TBI Study) of AD patients and non-demented controls. Group comparisons and correlation analyses of major ECM components in soluble and synaptosomal fractions from frontal, temporal cortex, and hippocampus of control, low-grade, and high-grade AD brains revealed a reduction in brevican in temporal cortex soluble and frontal cortex synaptosomal fractions in AD. In contrast, neurocan, aggrecan and the link protein HAPLN1 were up-regulated in soluble cortical fractions. In comparison, RNAseq data showed no correlation between aggrecan and brevican expression levels and Braak or CERAD stages, but for hippocampal expression of HAPLN1, neurocan and the brevican-interaction partner tenascin-R negative correlations with Braak stages were detected. CSF levels of brevican and neurocan in patients positively correlated with age, total tau, p-Tau, neurofilament-L and Aβ1-40. Negative correlations were detected with the Aβ ratio and the IgG index. Altogether, our study reveals spatially segregated molecular rearrangements of the ECM in AD brains at RNA or protein levels, which may contribute to the pathogenic process.
Collapse
Affiliation(s)
- Lukas Höhn
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Wilhelm Hußler
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Anni Richter
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, 07743 Jena, Germany
| | - Karl-Heinz Smalla
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39104 Magdeburg, Germany
- Institute for Pharmacology and Toxicology, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Anna-Maria Birkl-Toeglhofer
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Christoph Birkl
- Department of Neuroradiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39104 Magdeburg, Germany
| | - Stefan L. Leber
- Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, 8036 Graz, Austria
| | - Eckart D. Gundelfinger
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39104 Magdeburg, Germany
- Institute for Pharmacology and Toxicology, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39104 Magdeburg, Germany
- German Center for Neurodegenerative Disorders (DZNE), 39120 Magdeburg, Germany
| | - Constanze I. Seidenbecher
- Leibniz Institute for Neurobiology (LIN), 39118 Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, 07743 Jena, Germany
- Center for Behavioral Brain Sciences (CBBS), 39104 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
56
|
Agrawal N, Farhat NY, Sinaii N, Do AD, Xiao C, Berry-Kravis E, Bianconi S, Masvekar R, Bielekova B, Solomon B, Porter FD. Neurofilament light chain in cerebrospinal fluid as a novel biomarker in evaluating both clinical severity and therapeutic response in Niemann-Pick disease type C1. Genet Med 2023; 25:100349. [PMID: 36470574 PMCID: PMC9992339 DOI: 10.1016/j.gim.2022.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Niemann-Pick disease type C1 (NPC1) is a neurodegenerative lysosomal disorder caused by pathogenic variants in NPC1. Disease progression is monitored using the NPC Neurological Severity Scale, but there are currently no established validated or qualified biomarkers. Neurofilament light chain (NfL) is being investigated as a biomarker in multiple neurodegenerative diseases. METHODS Cross-sectional and longitudinal cerebrospinal fluid (CSF) samples were obtained from 116 individuals with NPC1. NfL levels were measured using a solid-phase sandwich enzyme-linked immunosorbent assay and compared with age-appropriate non-NPC1 comparison samples. RESULTS Median levels of NfL were elevated at baseline (1152 [680-1840] pg/mL) in NPC1 compared with controls (167 [82-372] pg/mL; P < .001). Elevated NfL levels were associated with more severe disease as assessed by both the 17-domain and 5-domain NPC Neurological Severity Score. Associations were also observed with ambulation, fine motor, speech, and swallowing scores. Although treatment with the investigational drug 2-hydroxypropyl-β-cyclodextrin (adrabetadex) did not decrease CSF NfL levels, miglustat therapy over time was associated with a decrease (odds ratio = 0.77, 95% CI = 0.62-0.96). CONCLUSION CSF NfL levels are increased in individuals with NPC1, associated with clinical disease severity, and decreased with miglustat therapy. These data suggest that NfL is a biomarker that may have utility in future therapeutic trials.
Collapse
Affiliation(s)
- Neena Agrawal
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Nicole Y Farhat
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, Clinical Center, National Institues of Health, Bethesda, MD
| | - An Dang Do
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Changrui Xiao
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Elizabeth Berry-Kravis
- Department of Neurological Sciences, Department of Pediatrics, Department of Biochemistry, Rush University Medical Center, Chicago, IL
| | - Simona Bianconi
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD; Southern California Permanente Medical Group, San Diego, CA
| | - Ruturaj Masvekar
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Bibiana Bielekova
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Beth Solomon
- Rehabilitaiton Medicine Department, Mark O. Hatfield Clinical Research Center, National Institutes of Health, Bethesda, MD
| | - Forbes D Porter
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
57
|
Tran KM, Kawauchi S, Kramár EA, Rezaie N, Liang HY, Sakr JS, Gomez-Arboledas A, Arreola MA, Cunha CD, Phan J, Wang S, Collins S, Walker A, Shi KX, Neumann J, Filimban G, Shi Z, Milinkeviciute G, Javonillo DI, Tran K, Gantuz M, Forner S, Swarup V, Tenner AJ, LaFerla FM, Wood MA, Mortazavi A, MacGregor GR, Green KN. A Trem2 R47H mouse model without cryptic splicing drives age- and disease-dependent tissue damage and synaptic loss in response to plaques. Mol Neurodegener 2023; 18:12. [PMID: 36803190 PMCID: PMC9938579 DOI: 10.1186/s13024-023-00598-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/19/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND The TREM2 R47H variant is one of the strongest genetic risk factors for late-onset Alzheimer's Disease (AD). Unfortunately, many current Trem2 R47H mouse models are associated with cryptic mRNA splicing of the mutant allele that produces a confounding reduction in protein product. To overcome this issue, we developed the Trem2R47H NSS (Normal Splice Site) mouse model in which the Trem2 allele is expressed at a similar level to the wild-type Trem2 allele without evidence of cryptic splicing products. METHODS Trem2R47H NSS mice were treated with the demyelinating agent cuprizone, or crossed with the 5xFAD mouse model of amyloidosis, to explore the impact of the TREM2 R47H variant on inflammatory responses to demyelination, plaque development, and the brain's response to plaques. RESULTS Trem2R47H NSS mice display an appropriate inflammatory response to cuprizone challenge, and do not recapitulate the null allele in terms of impeded inflammatory responses to demyelination. Utilizing the 5xFAD mouse model, we report age- and disease-dependent changes in Trem2R47H NSS mice in response to development of AD-like pathology. At an early (4-month-old) disease stage, hemizygous 5xFAD/homozygous Trem2R47H NSS (5xFAD/Trem2R47H NSS) mice have reduced size and number of microglia that display impaired interaction with plaques compared to microglia in age-matched 5xFAD hemizygous controls. This is associated with a suppressed inflammatory response but increased dystrophic neurites and axonal damage as measured by plasma neurofilament light chain (NfL) level. Homozygosity for Trem2R47H NSS suppressed LTP deficits and loss of presynaptic puncta caused by the 5xFAD transgene array in 4-month-old mice. At a more advanced (12-month-old) disease stage 5xFAD/Trem2R47H NSS mice no longer display impaired plaque-microglia interaction or suppressed inflammatory gene expression, although NfL levels remain elevated, and a unique interferon-related gene expression signature is seen. Twelve-month old Trem2R47H NSS mice also display LTP deficits and postsynaptic loss. CONCLUSIONS The Trem2R47H NSS mouse is a valuable model that can be used to investigate age-dependent effects of the AD-risk R47H mutation on TREM2 and microglial function including its effects on plaque development, microglial-plaque interaction, production of a unique interferon signature and associated tissue damage.
Collapse
Affiliation(s)
- Kristine M. Tran
- Department of Neurobiology and Behavior, University of California, Irvine, USA
| | - Shimako Kawauchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Enikö A. Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, USA
| | - Narges Rezaie
- Department of Developmental and Cell Biology, University of California, Irvine, USA
- Center for Complex Biological Systems, Irvine, USA
| | - Heidi Yahan Liang
- Department of Developmental and Cell Biology, University of California, Irvine, USA
- Center for Complex Biological Systems, Irvine, USA
| | - Jasmine S. Sakr
- Department of Pharmaceutical Sciences, University of California, Irvine, USA
| | | | - Miguel A. Arreola
- Department of Neurobiology and Behavior, University of California, Irvine, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Shuling Wang
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Sherilyn Collins
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Amber Walker
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Kai-Xuan Shi
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
| | - Ghassan Filimban
- Department of Developmental and Cell Biology, University of California, Irvine, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, USA
| | - Giedre Milinkeviciute
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Dominic I. Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Katelynn Tran
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Magdalena Gantuz
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, USA
- Center for Complex Biological Systems, Irvine, USA
| | - Andrea J. Tenner
- Department of Neurobiology and Behavior, University of California, Irvine, USA
- Department of Molecular Biology & Biochemistry, University of California, Irvine, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, USA
| | - Frank M. LaFerla
- Department of Neurobiology and Behavior, University of California, Irvine, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California, Irvine, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, USA
- Center for Complex Biological Systems, Irvine, USA
| | - Grant R. MacGregor
- Transgenic Mouse Facility, Office of Research, ULAR, Irvine, USA
- Department of Developmental and Cell Biology, University of California, Irvine, USA
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, USA
| |
Collapse
|
58
|
Ciardullo S, Muraca E, Bianconi E, Cannistraci R, Perra S, Zerbini F, Perseghin G. Diabetes Mellitus is Associated With Higher Serum Neurofilament Light Chain Levels in the General US Population. J Clin Endocrinol Metab 2023; 108:361-367. [PMID: 36196647 DOI: 10.1210/clinem/dgac580] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/09/2022] [Indexed: 01/20/2023]
Abstract
CONTEXT Serum neurofilament light chain (sNfL) levels are biomarkers of neuroaxonal injury in multiple neurological diseases. OBJECTIVE Given the paucity of data on the distribution of sNfL levels in the general population, in the present study we identified predictors of sNfL levels in a community setting and investigated the association between diabetes and sNfL. METHODS sNfL levels were measured in 2070 people aged 20 to 75 years from the general US population (275 with and 1795 without diabetes) that participated in the 2013-2014 cycle of the National Health and Nutrition Examination Survey. We evaluated the association between diabetes and sNfL levels after adjustment for age, sex, race-ethnicity, alcohol use, and kidney function using a multivariable linear regression model. Cognitive function was evaluated in a subset of participants aged 60 to 75 years using the Consortium to Establish a Registry for Alzheimer's Disease-Word Learning test, the Animal Fluency test, and the Digit Symbol Substitution test. RESULTS The weighted prevalence of diabetes was 10.4% (95% CI, 9.0-11.9). In each age stratum, patients with diabetes exhibited higher sNfL levels compared with nondiabetic participants. Age, proportion of males, prevalence of diabetes, and homeostatic model of insulin resistance increased progressively across quartiles of sNfL levels in the overall population, whereas estimated glomerular filtration rate (eGFR) showed an opposite trend. In the multivariable model, age, sex, eGFR, alcohol use and diabetes were significantly associated with sNfL levels. Moreover, higher sNfL levels were associated with worse performance in all 3 cognitive function tests. CONCLUSION Diabetes is associated with higher sNfL. Further large-scale and prospective studies are needed to replicate our results and evaluate the ability of sNfL to predict the incidence of neuropathy and dementia in this patient population.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
| | - Emanuele Muraca
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
| | - Eleonora Bianconi
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
| | - Rosa Cannistraci
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
| | - Silvia Perra
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
| | - Francesca Zerbini
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Policlinico di Monza, 20900 Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
| |
Collapse
|
59
|
Zhang Q, Fan W, Sun J, Zhang J, Yin Y. Review of Neurofilaments as Biomarkers in Sepsis-Associated Encephalopathy. J Inflamm Res 2023; 16:161-168. [PMID: 36660377 PMCID: PMC9843472 DOI: 10.2147/jir.s391325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/24/2022] [Indexed: 01/12/2023] Open
Abstract
Sepsis is a common and fatal disease, especially in critically ill patients. Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction with acute altered consciousness, permanent cognitive impairment, and even coma, accompanied by sepsis, without direct central nervous system infection. When managing SAE, early identification and quantification of axonal damage facilitate faster and more accurate diagnosis and prognosis. Although no specific markers for SAE have been identified, several biomarkers have been proposed. Neurofilament light chain (NFL) is a highly expressed cytoskeletal component of neurofilament (NF) proteins that can be found in blood and cerebrospinal fluid (CSF) after exposure to axonal injury. NFs can be used as diagnostic and prognostic biomarkers for sepsis-related brain injury. Phosphorylation of NFs contributes to the maturation and stabilization of cytoskeletal structures, especially axons, and facilitates axonal transport, including mitochondrial transport and energy transport. The stability of NF proteins can be assessed by monitoring the expression of NF genes. Furthermore, phosphorylation levels of NFs can be monitored to determine mitochondrial axonal transport associated with cellular energy metabolism at distal axons to assess progression during SAE treatment. This paper provides new insights into the biological characteristics, detection techniques, and scientific achievements of NFs, and discusses the underlying mechanisms and future research directions of NFs in SAE.
Collapse
Affiliation(s)
- Qiulei Zhang
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Weixuan Fan
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Jian Sun
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingxiao Zhang
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China,Correspondence: Jingxiao Zhang; Yongjie Yin, Tel +86-13756314698; +86-13596103459, Email ;
| | - Yongjie Yin
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
60
|
CSF1R inhibitors induce a sex-specific resilient microglial phenotype and functional rescue in a tauopathy mouse model. Nat Commun 2023; 14:118. [PMID: 36624100 PMCID: PMC9829908 DOI: 10.1038/s41467-022-35753-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.
Collapse
|
61
|
Li Y, Zheng JJ, Wu X, Gao W, Liu CJ. Postural control of Parkinson's disease: A visualized analysis based on Citespace knowledge graph. Front Aging Neurosci 2023; 15:1136177. [PMID: 37032828 PMCID: PMC10080997 DOI: 10.3389/fnagi.2023.1136177] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
Postural control impairment is one of the primary motor symptoms in patients with Parkinson's disease, leading to an increased risk of falling. Several studies have been conducted on postural control disorders in Parkinson's disease patients, but no relevant bibliometric analysis has been found. In this paper, the Web of Science Core Collection database was searched for 1,295 relevant papers on postural control in Parkinson's disease patients from December 2011 to December 2021. Based on the Citespace knowledge graph, these relevant papers over the last decade were analyzed from the perspectives of annual publication volume, countries and institutes cooperation, authors cooperation, dual-map overlay of journals, co-citation literature, and keywords. The purpose of this study was to explore the current research status, research hotspots, and frontiers in this field, and to provide a reference for further promoting the research on postural control in Parkinson's disease patients.
Collapse
Affiliation(s)
- Yan Li
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Jie-Jiao Zheng
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Shanghai Clinical Research Center for Rehabilitation Medicine, Shanghai, China
- *Correspondence: Jie-Jiao Zheng,
| | - Xie Wu
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Wen Gao
- Department of Rehabilitation Medicine, Huadong Hospital, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Rehabilitation Medicine, Shanghai, China
| | - Chan-Jing Liu
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
62
|
Shim Y. Follow-up Comparisons of Two Plasma Biomarkers of Alzheimer's Disease, Neurofilament Light Chain, and Oligomeric Aβ: A Pilot Study. Curr Alzheimer Res 2023; 20:715-724. [PMID: 38299421 DOI: 10.2174/0115672050284054240119101834] [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: 11/19/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND AND OBJECTIVE Recent evidence suggests that blood-based biomarkers might be useful for Alzheimer's disease (AD). Among them, we intend to investigate whether neurofilament light (NfL) and multimer detection system-oligomeric Aβ (MDS-OAβ) values can be useful in screening, predicting, and monitoring disease progression and how the relationship between NfL and MDS-OAβ values changes. METHODS Eighty participants with probable AD dementia, 50 with mild cognitive impairment (MCI), and 19 with subjective cognitive decline (SCD) underwent baseline and follow-up evaluations of the Mini-Mental Status Examination (MMSE) and both plasma biomarkers. RESULTS Baseline MDS-OAß (p = 0.016) and NfL (p = 0.002) plasma concentrations differed significantly among groups, but only NfL correlated with baseline MMSE scores (r = -0.278, p = 0.001). In follow-up, neither correlated with MMSE changes overall. However, in SCD and MCI participants (n = 32), baseline MDS-OAß correlated with follow-up MMSE scores (r = 0.532, p = 0.041). Linear regression revealed a relationship between baseline MDS-OAβ and follow-up MMSE scores. In SCD and MCI participants, plasma NfL changes correlated with MMSE changes (r = 0.564, p = 0.028). CONCLUSION This study shows that only in participants with SCD and MCI, not including AD dementia, can MDS-OAß predict the longitudinal cognitive decline measured by follow-up MMSE. Changes of NfL, not MDS-OAß, parallel the changes of MMSE. Further studies with larger samples and longer durations could strengthen these results..
Collapse
Affiliation(s)
- YongSoo Shim
- Department of Neurology, The Catholic University of Korea Eunpyeong St. Mary's Hospital, Seoul, Republic of Korea
| |
Collapse
|
63
|
Götze K, Vrillon A, Bouaziz-Amar E, Mouton-Liger F, Hugon J, Martinet M, Dumurgier J, Cognat E, Zetterberg H, Blennow K, Hourrègue C, Paquet C, Lilamand M. Plasma neurofilament light chain in memory clinic practice: Evidence from a real-life study. Neurobiol Dis 2023; 176:105937. [PMID: 36462720 DOI: 10.1016/j.nbd.2022.105937] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE To explore the accuracy of plasma neurofilament light chain (NfL) as a biomarker for diagnosis and staging of cognitive impairment, in a large cohort with of previously diagnosed patients in clinical practice. METHODS Retrospective, cross-sectional, monocentric study, from a tertiary memory clinic. Patients underwent cerebrospinal fluid core Alzheimer's disease (AD) biomarker evaluation using ELISA or Elecsys methods, and plasma NfL analysis using the single molecule array technology. The patients' biomarker data were examined for associations with: i/cognitive status ii/presence of neurodegenerative disease and iii/diagnostic groups. Associations between core CSF biomarkers and plasma NfL were determined. RESULTS Participants (N = 558, mean age = 69.2 ± 8.8, 56.5% women) were diagnosed with AD (n = 274, considering dementia and MCI stages), frontotemporal dementia (FTD, n = 55), Lewy body disease (LBD, n = 40, considering MCI and dementia stages), other neurodegenerative diseases, n = 57 (e.g Supranuclear Palsy, Corticobasal syndrome), non-neurodegenerative cognitive impairment (NND, n = 79, e.g. vascular lesions, epilepsy or psychiatric disorders) or subjective cognitive impairment (SCI, n = 53). Mean plasma NfL (log, pg/mL) levels were higher in neurodegenerative than non-neurodegenerative disorders (1.35 ± 0.2 vs 1.16 ± 0.23, p < 0.001), higher in all diagnostic groups than in SCI (1.06 ± 0.23) p < 0.001), and associated with the stage of cognitive impairment (p < 0.001). The addition of plasma NfL to a clinical model (age, MMSE and APOE ε4 carriership) marginally improved the discrimination of degenerative from non-degenerative disorders in ROC analysis (AUC clinical model: 0.81, 95% CI = [0.77;0.85] AUC clinical model + plasma NfL: AUC = 0.83 95% CI = [0.78;0.87], delta Akaike information criterion = -11.7). DISCUSSION Plasma NfL could help discrimination between degenerative and non-degenerative cognitive disorders, albeit not better than comprehensive clinical evaluation.
Collapse
Affiliation(s)
- Karl Götze
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France.
| | - Agathe Vrillon
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France
| | - Elodie Bouaziz-Amar
- Inserm Unit UMR S-1144, Paris, France; Biochemistery Department, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France
| | | | - Jacques Hugon
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France
| | | | - Julien Dumurgier
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France
| | - Emmanuel Cognat
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Claire Hourrègue
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France
| | - Claire Paquet
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France
| | - Matthieu Lilamand
- Cognitive Neurology Center, Lariboisière Hospital (GHU AP-HP.Nord), Paris, France; Inserm Unit UMR S-1144, Paris, France
| |
Collapse
|
64
|
Pagonabarraga J, Pérez-González R, Bejr-Kasem H, Marín-Lahoz J, Horta-Barba A, Martinez-Horta S, Aracil-Bolaños I, Sampedro F, Campolongo A, Rivas E, Puig-Davi A, Ruiz-Barrios I, Pérez-Pérez J, Pascual-Sedano B, Kulisevsky J. Dissociable contribution of plasma NfL and p-tau181 to cognitive impairment in Parkinson's disease. Parkinsonism Relat Disord 2022; 105:132-138. [PMID: 35752549 DOI: 10.1016/j.parkreldis.2022.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cognitive dysfunction is a disabling complication in Parkinson's disease (PD). Accuracy of diagnosis of mild cognitive impairment in PD (PD-MCI) depends on the tests performed, which limits results generalization. Blood-based biomarkers could provide additional objective information for PD-MCI diagnosis and progression. Blood neurofilament light chain (NfL), a marker of neuronal injury, has shown good performance for PD disease stratification and progression. While NfL is not disease-specific, phosphorylated-tau at threonine-181 (p-tau181) in blood is a highly specific marker of concomitant brain amyloid-β and tau pathology. METHODS We investigated the potential of plasma NfL and p-tau181 levels as markers of cognitive impairment in a prospective cohort of 109 PD patients with and without PD-MCI (age 68.1 ± 7 years, education 12.2± 5 years), and 40 comparable healthy controls. After a follow-up of 4 years, we evaluated their predictive value for progression to dementia. RESULTS Although NfL and p-tau181 levels were significantly increased in PD compared with healthy controls, only NfL levels were significantly higher in PD-MCI compared with PD with normal cognition (PD-NC) at baseline. After a follow-up of 4 years, only NfL predicted progression to dementia (HR 1.23, 95% CI 1.02-1.53; p = 0.038). Significant correlations between fluid biomarkers and neuropsychological examination were only found with NfL levels. CONCLUSIONS Plasma NfL levels objectively differentiates PD-MCI from PD-NC patients, and may serve as a plasma biomarker for predicting progression to dementia in PD. Plasma levels of p-tau181 does not seem to help in differentiating PD-MCI or to predict future cognitive deterioration.
Collapse
Affiliation(s)
- Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Rocío Pérez-González
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Helena Bejr-Kasem
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Juan Marín-Lahoz
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Saul Martinez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Ignacio Aracil-Bolaños
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Frederic Sampedro
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Antonia Campolongo
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Elisa Rivas
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Arnau Puig-Davi
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - I Ruiz-Barrios
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain
| | - Jesús Pérez-Pérez
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Berta Pascual-Sedano
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Department of Medicine, Barcelona, Spain; Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
65
|
Rother C, Uhlmann RE, Müller SA, Schelle J, Skodras A, Obermüller U, Häsler LM, Lambert M, Baumann F, Xu Y, Bergmann C, Salvadori G, Loos M, Brzak I, Shimshek D, Neumann U, Walker LC, Schultz SA, Chhatwal JP, Kaeser SA, Lichtenthaler SF, Staufenbiel M, Jucker M. Experimental evidence for temporal uncoupling of brain Aβ deposition and neurodegenerative sequelae. Nat Commun 2022; 13:7333. [PMID: 36443293 PMCID: PMC9705543 DOI: 10.1038/s41467-022-34538-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Brain Aβ deposition is a key early event in the pathogenesis of Alzheimer´s disease (AD), but the long presymptomatic phase and poor correlation between Aβ deposition and clinical symptoms remain puzzling. To elucidate the dependency of downstream pathologies on Aβ, we analyzed the trajectories of cerebral Aβ accumulation, Aβ seeding activity, and neurofilament light chain (NfL) in the CSF (a biomarker of neurodegeneration) in Aβ-precursor protein transgenic mice. We find that Aβ deposition increases linearly until it reaches an apparent plateau at a late age, while Aβ seeding activity increases more rapidly and reaches a plateau earlier, coinciding with the onset of a robust increase of CSF NfL. Short-term inhibition of Aβ generation in amyloid-laden mice reduced Aβ deposition and associated glial changes, but failed to reduce Aβ seeding activity, and CSF NfL continued to increase although at a slower pace. When short-term or long-term inhibition of Aβ generation was started at pre-amyloid stages, CSF NfL did not increase despite some Aβ deposition, microglial activation, and robust brain Aβ seeding activity. A dissociation of Aβ load and CSF NfL trajectories was also found in familial AD, consistent with the view that Aβ aggregation is not kinetically coupled to neurotoxicity. Rather, neurodegeneration starts when Aβ seeding activity is saturated and before Aβ deposition reaches critical (half-maximal) levels, a phenomenon reminiscent of the two pathogenic phases in prion disease.
Collapse
Affiliation(s)
- Christine Rother
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Ruth E Uhlmann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Juliane Schelle
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Angelos Skodras
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Ulrike Obermüller
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Lisa M Häsler
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Frank Baumann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Ying Xu
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Carina Bergmann
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Giulia Salvadori
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Maarten Loos
- Sylics (Synaptologics BV), 3721 MA, Bilthoven, The Netherlands
| | - Irena Brzak
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Derya Shimshek
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Ulf Neumann
- Novartis Institutes for Biomedical Research, CH-4056, Basel, Switzerland
| | - Lary C Walker
- Department of Neurology and Emory National Primate Research Center, Emory University, Atlanta, GA, 30322, USA
| | | | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Stephan A Kaeser
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Staufenbiel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, D-72076, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
66
|
Simoes FA, Joilin G, Peters O, Schneider LS, Priller J, Spruth EJ, Vogt I, Kimmich O, Spottke A, Hoffmann DC, Falkenburger B, Brandt M, Prudlo J, Brockmann K, Fries FL, Rowe JB, Church A, Respondek G, Newbury SF, Leigh PN, Morris HR, Höglinger GU, Hafezparast M. Potential of Non-Coding RNA as Biomarkers for Progressive Supranuclear Palsy. Int J Mol Sci 2022; 23:ijms232314554. [PMID: 36498882 PMCID: PMC9738832 DOI: 10.3390/ijms232314554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Objective markers for the neurodegenerative disorder progressive supranuclear palsy (PSP) are needed to provide a timely diagnosis with greater certainty. Non-coding RNA (ncRNA), including microRNA, piwi-interacting RNA, and transfer RNA, are good candidate markers in other neurodegenerative diseases, but have not been investigated in PSP. Therefore, as proof of principle, we sought to identify whether they were dysregulated in matched serum and cerebrospinal fluid (CSF) samples of patients with PSP. Small RNA-seq was undertaken on serum and CSF samples from healthy controls (n = 20) and patients with PSP (n = 31) in two cohorts, with reverse transcription-quantitative PCR (RT-qPCR) to confirm their dysregulation. Using RT-qPCR, we found in serum significant down-regulation in hsa-miR-92a-3p, hsa-miR-626, hsa-piR-31068, and tRNA-ValCAC. In CSF, both hsa-let-7a-5p and hsa-piR-31068 showed significant up-regulation, consistent with their changes observed in the RNA-seq results. Interestingly, we saw no correlation in the expression of hsa-piR-31068 within our matched serum and CSF samples, suggesting there is no common dysregulatory mechanism between the two biofluids. While these changes were in a small cohort of samples, we have provided novel evidence that ncRNA in biofluids could be possible diagnostic biomarkers for PSP and further work will help to expand this potential.
Collapse
Affiliation(s)
- Fabio A. Simoes
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Greig Joilin
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany
| | | | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Psychiatry and Psychotherapy, Charité, 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Psychiatry and Psychotherapy, Charité, 10117 Berlin, Germany
| | - Ina Vogt
- German Center for Neurodegenerative Diseases (DZNE), Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, University of Bonn, Bonn 53127, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, University of Bonn, Bonn 53127, Germany
| | | | - Björn Falkenburger
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Moritz Brandt
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Johannes Prudlo
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Franca Laura Fries
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - James B. Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge CB2 0QQ, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK
| | - Alistair Church
- Department of Neurology, Royal Gwent Hospital, Newport NP20 2UB, UK
| | - Gesine Respondek
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, Technische Universität München, 81377 Munich, Germany
| | | | - P. Nigel Leigh
- Brighton and Sussex Medical School, Brighton BN1 9QG, UK
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Günter U. Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Germany
- Department of Neurology, Technische Universität München, 81377 Munich, Germany
| | - Majid Hafezparast
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
- Correspondence: ; Tel.: +44-1273-678214
| |
Collapse
|
67
|
Liu C, Guo X, Si H, Li G. A mink (Neovison vison) model of self-injury: Effects of CBP-CREB axis on neuronal damage and behavior. Front Vet Sci 2022; 9:975112. [DOI: 10.3389/fvets.2022.975112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
ObjectiveSelf-injurious behavior (SIB) is a clinically challenging problem in the general population and several clinical disorders. However, the precise molecular mechanism of SIB is still not clear. In this paper, the systematic investigation of the genesis and development of SIB is conducted based on behavioral and pathophysiology studies in mink (Neovison vison) models.MethodThe night-vision video was used to observe the mink behavior, and the duration was a month. HE stain was performed to characterize the pathology change in the brain of a mink. IHC assay was performed to conduct the protein level detection of Iba-1, p-CREB, CBP, and p300 in the brain tissues. Elisa assay was used to examine the levels of NfL and NfH in serum and CSF of mink. The qRT-PCR assay was used to detect the expression of Bcl-2, NOR1, FoxO4, c-FOS, CBP, and p300 in brain tissues. Western blot was used to detect the protein levels of p-CREB, CBP, and p300 in brain tissues. We also used Evans Blue as a tracer to detect whether the blood-brain barrier was impaired in the brain of mink.ResultThe behavioral test, histopathological and molecular biology experiments were combined in this paper, and the results showed that CBP was related to SIB. Mechanism analysis showed that the dysregulation of CBP in brain-activated CREB signaling will result in nerve damage of the brain and SIB symptoms in minks. More importantly, the CBP-CREB interaction inhibitor might help relieve SIB and nerve damage in brain tissues.ConclusionOur results illustrate that the induction of CBP and the activation of CREB are novel mechanisms in the genesis of SIB. This finding indicates that the CBP-CREB axis is critical for SIB and demonstrates the efficacy of the CBP-CREB interaction inhibitor in treating these behaviors.
Collapse
|
68
|
Yeo T, Bayuangga H, Augusto-Oliveira M, Sealey M, Claridge TDW, Tanner R, Leppert D, Palace J, Kuhle J, Probert F, Anthony DC. Metabolomics detects clinically silent neuroinflammatory lesions earlier than neurofilament-light chain in a focal multiple sclerosis animal model. J Neuroinflammation 2022; 19:252. [PMID: 36210459 PMCID: PMC9549622 DOI: 10.1186/s12974-022-02614-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Background Despite widespread searches, there are currently no validated biofluid markers for the detection of subclinical neuroinflammation in multiple sclerosis (MS). The dynamic nature of human metabolism in response to changes in homeostasis, as measured by metabolomics, may allow early identification of clinically silent neuroinflammation. Using the delayed-type hypersensitivity (DTH) MS rat model, we investigated the serum and cerebrospinal fluid (CSF) metabolomics profiles and neurofilament-light chain (NfL) levels, as a putative marker of neuroaxonal damage, arising from focal, clinically silent neuroinflammatory brain lesions and their discriminatory abilities to distinguish DTH animals from controls. Methods 1H nuclear magnetic resonance (NMR) spectroscopy metabolomics and NfL measurements were performed on serum and CSF at days 12, 28 and 60 after DTH lesion initiation. Supervised multivariate analyses were used to determine metabolomics differences between DTH animals and controls. Immunohistochemistry was used to assess the extent of neuroinflammation and tissue damage. Results Serum and CSF metabolomics perturbations were detectable in DTH animals (vs. controls) at all time points, with the greatest change occurring at the earliest time point (day 12) when the neuroinflammatory response was most intense (mean predictive accuracy [SD]—serum: 80.6 [10.7]%, p < 0.0001; CSF: 69.3 [13.5]%, p < 0.0001). The top discriminatory metabolites at day 12 (serum: allantoin, cytidine; CSF: glutamine, glucose) were all reduced in DTH animals compared to controls, and correlated with histological markers of neuroinflammation, particularly astrogliosis (Pearson coefficient, r—allantoin: r = − 0.562, p = 0.004; glutamine: r = − 0.528, p = 0.008). Serum and CSF NfL levels did not distinguish DTH animals from controls at day 12, rather, significant differences were observed at day 28 (mean [SEM]—serum: 38.5 [4.8] vs. 17.4 [2.6] pg/mL, p = 0.002; CSF: 1312.0 [379.1] vs. 475.8 [74.7] pg/mL, p = 0.027). Neither serum nor CSF NfL levels correlated with markers of neuroinflammation; serum NfL did, however, correlate strongly with axonal loss (r = 0.641, p = 0.001), but CSF NfL did not (p = 0.137). Conclusions While NfL levels were elevated later in the pathogenesis of the DTH lesion, serum and CSF metabolomics were able to detect early, clinically silent neuroinflammation and are likely to present sensitive biomarkers for the assessment of subclinical disease activity in patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02614-8.
Collapse
|
69
|
Liu Y, Dou K, Xue L, Li X, Xie A. Neurofilament light as a biomarker for motor decline in Parkinson’s disease. Front Neurosci 2022; 16:959261. [PMID: 36117629 PMCID: PMC9477093 DOI: 10.3389/fnins.2022.959261] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/05/2022] [Indexed: 11/15/2022] Open
Abstract
Objectives The aim of this study was to determine whether neurofifilament light (NfL) could reflect motor decline and compare the predictive values of cerebrospinal fluid (CSF) and serum NfL in individuals with PD. Methods CSF/serum samples were collected from patients with PD and healthy controls (HCs) with motor assessments at baseline and after three years of follow-up from the Parkinson’s Progression Markers Initiative (PPMI). Multiple linear regression models and linear mixed-effects models were used to investigate the associations of motor assessments with baseline and longitudinal CSF/serum NfL. Associations between the change rates of motor assessments and CSF/serum NfL were further investigated via multiple linear regression models. Mediating effect analysis was used to research whether CSF alpha-synuclein (α-syn) acts as the mediator between NfL and motor assessments. Results We found patients with PD had higher baseline CSF/serum NfL levels than HCs. Both baseline CSF/serum NfLs and their change rates predicted measurable motor decline in PD assessed by different motor scores. Baseline serum NfL and its rate of change were strongly associated with CSF NfL levels in patients with PD (P < 0.001). Besides, there were also significant differences in CSF/serum NfL levels and predicted values of motor decline between men and women with PD. Mediating effect analysis showed CSF α-syn mediated the effect of CSF NfL on total Unified Parkinson’s Disease Rating Scale (UPDRS) scores and UPDRSIII with 30.6 and 20.2% mediation, respectively. Conclusion Our results indicated that NfL, especially serum NfL concentration, could serve as an easily accessible biomarker to monitor the severity and progression of motor decline in individuals with PD, especially in men with PD. Besides, CSF α-syn acts as a mediator between NfL and motor progression.
Collapse
Affiliation(s)
- Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kaixin Dou
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ling Xue
- Department of Nursing, Tai’an City Central Hospital, Tai’an, China
| | - Xiaoyuan Li
- Department of Traditional Chinese Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Anmu Xie,
| |
Collapse
|
70
|
Kölliker Frers RA, Otero-Losada M, Kobiec T, Udovin LD, Aon Bertolino ML, Herrera MI, Capani F. Multidimensional overview of neurofilament light chain contribution to comprehensively understanding multiple sclerosis. Front Immunol 2022; 13:912005. [PMID: 35967312 PMCID: PMC9368191 DOI: 10.3389/fimmu.2022.912005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease characterized by demyelination, progressive axonal loss, and varying clinical presentations. Axonal damage associated with the inflammatory process causes neurofilaments, the major neuron structural proteins, to be released into the extracellular space, reaching the cerebrospinal fluid (CSF) and the peripheral blood. Methodological advances in neurofilaments’ serological detection and imaging technology, along with many clinical and therapeutic studies in the last years, have deepened our understanding of MS immunopathogenesis. This review examines the use of light chain neurofilaments (NFLs) as peripheral MS biomarkers in light of the current clinical and therapeutic evidence, MS immunopathology, and technological advances in diagnostic tools. It aims to highlight NFL multidimensional value as a reliable MS biomarker with a diagnostic-prognostic profile while improving our comprehension of inflammatory neurodegenerative processes, mainly RRMS, the most frequent clinical presentation of MS.
Collapse
Affiliation(s)
- Rodolfo A. Kölliker Frers
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Unidad de Parasitología, Hospital J. M. Ramos Mejía, Buenos Aires, Argentina
| | - Matilde Otero-Losada
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- *Correspondence: Matilde Otero-Losada,
| | - Tamara Kobiec
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Lucas D. Udovin
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
| | - María Laura Aon Bertolino
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
| | - María I. Herrera
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Francisco Capani
-
Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas (CAECIHS. UAI-CONICET), Buenos Aires, Argentina
- Departamento de Biología, Universidad Argentina John Kennedy (UAJK), Buenos Aires, Argentina
| |
Collapse
|
71
|
Comparative Analysis of Neurodegeneration and Axonal Dysfunction Biomarkers in the Cerebrospinal Fluid of Patients with Multiple Sclerosis. J Clin Med 2022; 11:jcm11144122. [PMID: 35887886 PMCID: PMC9324050 DOI: 10.3390/jcm11144122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Given the significant role of neurodegeneration in the progression of multiple sclerosis (MS) and insufficient therapies, there is an urgent need to better understand this pathology and to find new biomarkers that could provide important insight into the biological mechanisms of the disease. Thus, the present study aimed to compare different neurodegeneration and axonal dysfunction biomarkers in MS and verify their potential clinical usefulness. METHODS A total of 59 patients, who underwent CSF analysis during their diagnostics, were enrolled in the study. Quantitative analysis of neurodegeneration biomarkers was performed through immunological tests. Oligoclonal bands were detected by isoelectric focusing on agarose gel, whereas the concentrations of immunoglobulins and albumin were measured using nephelometry. RESULTS Our studies showed that NfL, RTN4, and tau protein enabled the differentiation of MS patients from the control group. Additionally, the baseline CSF NfL levels positively correlated with the tau and MRI results, whereas the RTN4 concentrations were associated with the immunoglobulin quotients. The AUC for NfL was the highest among the tested proteins, although the DeLong test of the ROC curves showed no significant difference between the AUCs for NfL and RTN4. CONCLUSION The CSF NfL, RTN-4, and tau levels at the time of diagnosis could be potential diagnostic markers of multiple sclerosis, although NfL seems to have the best clinical value.
Collapse
|
72
|
Wai CH, Jin J, Cyrklaff M, Genoud C, Funaya C, Sattler J, Maceski A, Meier S, Heiland S, Lanzer M, Frischknecht F, Kuhle J, Bendszus M, Hoffmann A. Neurofilament light chain plasma levels are associated with area of brain damage in experimental cerebral malaria. Sci Rep 2022; 12:10726. [PMID: 35750882 PMCID: PMC9232608 DOI: 10.1038/s41598-022-14291-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Abstract
Neurofilament light chain (NfL), released during central nervous injury, has evolved as a powerful serum marker of disease severity in many neurological disorders, including infectious diseases. So far NfL has not been assessed in cerebral malaria in human or its rodent model experimental cerebral malaria (ECM), a disease that can lead to fatal brain edema or reversible brain edema. In this study we assessed if NfL serum levels can also grade disease severity in an ECM mouse model with reversible (n = 11) and irreversible edema (n = 10). Blood–brain-barrier disruption and brain volume were determined by magnetic resonance imaging. Neurofilament density volume as well as structural integrity were examined by electron microscopy in regions of most severe brain damage (olfactory bulb (OB), cortex and brainstem). NfL plasma levels in mice with irreversible edema (317.0 ± 45.01 pg/ml) or reversible edema (528.3 ± 125.4 pg/ml) were significantly increased compared to controls (103.4 ± 25.78 pg/ml) by three to five fold, but did not differ significantly in mice with reversible or irreversible edema. In both reversible and irreversible edema, the brain region most affected was the OB with highest level of blood–brain-barrier disruption and most pronounced decrease in neurofilament density volume, which correlated with NfL plasma levels (r = − 0.68, p = 0.045). In cortical and brainstem regions neurofilament density was only decreased in mice with irreversible edema and strongest in the brainstem. In reversible edema NfL plasma levels, MRI findings and neurofilament volume density normalized at 3 months’ follow-up. In conclusion, NfL plasma levels are elevated during ECM confirming brain damage. However, NfL plasma levels fail short on reliably indicating on the final outcomes in the acute disease stage that could be either fatal or reversible. Increased levels of plasma NfL during the acute disease stage are thus likely driven by the anatomical location of brain damage, the olfactory bulb, a region that serves as cerebral draining pathway into the nasal lymphatics.
Collapse
Affiliation(s)
- Chi Ho Wai
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Jessica Jin
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Genoud
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Charlotta Funaya
- Electron Microscopy Core Facility, Heidelberg University, Heidelberg, Germany
| | - Julia Sattler
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Aleksandra Maceski
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephanie Meier
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Lanzer
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. .,Department of Neuroradiology, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Freiburgstrasse, 3010, Bern, Switzerland.
| |
Collapse
|
73
|
Wong YY, Wu CY, Yu D, Kim E, Wong M, Elez R, Zebarth J, Ouk M, Tan J, Liao J, Haydarian E, Li S, Fang Y, Li P, Pakosh M, Tartaglia MC, Masellis M, Swardfager W. Biofluid markers of blood-brain barrier disruption and neurodegeneration in Lewy body spectrum diseases: A systematic review and meta-analysis. Parkinsonism Relat Disord 2022; 101:119-128. [PMID: 35760718 DOI: 10.1016/j.parkreldis.2022.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mixed evidence supports blood-brain barrier (BBB) dysfunction in Lewy body spectrum diseases. METHODS We compare biofluid markers in people with idiopathic Parkinson's disease (PD) and people with PD dementia (PDD) and/or dementia with Lewy bodies (DLB), compared with healthy controls (HC). Seven databases were searched up to May 10, 2021. Outcomes included cerebrospinal fluid to blood albumin ratio (Qalb), and concentrations of 7 blood protein markers that also reflect BBB disruption and/or neurodegenerative co-pathology. We further explore differences between PD patients with and without evidence of dementia. Random-effects models were used to obtain standardized mean differences (SMD) with 95% confidence interval. RESULTS Of 13,949 unique records, 51 studies were meta-analyzed. Compared to HC, Qalb was higher in PD (NPD/NHC = 224/563; SMD = 0.960 [0.227-1.694], p = 0.010; I2 = 92.2%) and in PDD/DLB (NPDD/DLB/NHC = 265/670; SMD = 1.126 [0.358-1.893], p < 0.001; I2 = 78.2%). Blood neurofilament light chain (NfL) was higher in PD (NPD/NHC = 1848/1130; SMD = 0.747 [0.442-1.052], p < 0.001; I2 = 91.9%) and PDD/DLB (NPDD/DLB/NHC = 183/469; SMD = 1.051 [0.678-1.423], p = 0.004; I2 = 92.7%) than in HC. p-tau 181 (NPD/NHC = 276/164; SMD = 0.698 [0.149-1.247], p = 0.013; I2 = 82.7%) was also higher in PD compared to HC. In exploratory analyses, blood NfL was higher in PD without dementia (NPDND/NHC = 1005/740; SMD = 0.252 [0.042-0.462], p = 0.018; I2 = 71.8%) and higher in PDD (NPDD/NHC = 100/111; SMD = 0.780 [0.347-1.214], p < 0.001; I2 = 46.7%) compared to HC. Qalb (NPDD/NPDND = 63/191; SMD = 0.482 [0.189-0.774], p = 0.010; I2<0.001%) and NfL (NPDD/NPDND = 100/223; SMD = 0.595 [0.346-0.844], p < 0.001; I2 = 3.4%) were higher in PDD than in PD without dementia. CONCLUSIONS Biofluid markers suggest BBB disruption and neurodegenerative co-pathology involvement in common Lewy body diseases. Greater evidence of BBB breakdown was seen in Lewy body disease with cognitive impairment.
Collapse
Affiliation(s)
- Yuen Yan Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Di Yu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Esther Kim
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Renata Elez
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Zebarth
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Ouk
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jocelyn Tan
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jiamin Liao
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Eileen Haydarian
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Siming Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Yaolu Fang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Peihao Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Maureen Pakosh
- Library & Information Services, UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Mario Masellis
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada.
| |
Collapse
|
74
|
Hjartarson HT, Nathorst-Böös K, Sejersen T. Disease Modifying Therapies for the Management of Children with Spinal Muscular Atrophy (5q SMA): An Update on the Emerging Evidence. Drug Des Devel Ther 2022; 16:1865-1883. [PMID: 35734367 PMCID: PMC9208376 DOI: 10.2147/dddt.s214174] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
SMA (5q SMA) is an autosomal recessive neuromuscular disease with an estimated incidence of approximately 1 in 11,000 live births, characterized by progressive degeneration and loss of α-motor neurons in the spinal cord and brain stem, resulting in progressive muscle weakness. The disease spectrum is wide, from a serious congenital to a mild adult-onset disease. SMA is caused by biallelic mutations in the SMN1 gene and disease severity is modified primarily by SMN2 copy number. Before the advent of specific disease altering treatments, SMA was the second most common fatal autosomal recessive disorder after cystic fibrosis and the most common genetic cause of infant mortality. Nusinersen, risdiplam, and onasemnogene abeparvovec are presently the only approved disease modifying therapies for SMA, and the aim of this review is to discuss their mode of action, effects, safety concerns, and results from real-world experience. All exert their action by increasing the level of SMN protein in lower motor neuron. Nusinersen and risdiplam by modifying the SMN2 gene product, and onasemnogene abeparvovec by delivering SMN1 gene copies into cells. All have an established clinical efficacy. An important feature shared by all three is that early intervention is associated with a better treatment outcome, such that in cases where treatment is initiated in an early pre-symptomatic period, it may result in normal - or almost normal - motor development. Thus, early diagnosis followed by swift initiation of treatment is fundamental for the treatment response and consequently long-term prognosis in SMA type 1, and probably SMA type 2. The same principle similarly applies to the milder phenotypes. All three therapies are relatively novel, with risdiplam being the latest addition. Except for nusinersen, real-world data are still scarce, and long-term data are quite naturally lacking.
Collapse
Affiliation(s)
- Helgi Thor Hjartarson
- Department of Neuropediatrics, Astrid Lindgren Children´s Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Kristofer Nathorst-Böös
- Department of Neuropediatrics, Astrid Lindgren Children´s Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Sejersen
- Department of Neuropediatrics, Astrid Lindgren Children´s Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
75
|
Abstract
SignificanceSingle-cell transcriptomics has revealed specific glial activation states associated with the pathogenesis of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease (AD and PD). What is still needed are clinically relevant biomarkers for deciphering such glial states in AD and PD patients. To this end, we applied proteome analysis in cerebrospinal fluid (CSF) of mouse models of AD and PD pathology. This allowed us to identify a panel of glial CSF proteins that largely match the transcriptomic changes. The identified proteins can also be quantified in human CSF and show changes in AD patients, supporting their relevance as biomarker candidates to stage glial activation in patients with neurodegenerative diseases.
Collapse
|
76
|
Xia D, Lianoglou S, Sandmann T, Calvert M, Suh JH, Thomsen E, Dugas J, Pizzo ME, DeVos SL, Earr TK, Lin CC, Davis S, Ha C, Leung AWS, Nguyen H, Chau R, Yulyaningsih E, Lopez I, Solanoy H, Masoud ST, Liang CC, Lin K, Astarita G, Khoury N, Zuchero JY, Thorne RG, Shen K, Miller S, Palop JJ, Garceau D, Sasner M, Whitesell JD, Harris JA, Hummel S, Gnörich J, Wind K, Kunze L, Zatcepin A, Brendel M, Willem M, Haass C, Barnett D, Zimmer TS, Orr AG, Scearce-Levie K, Lewcock JW, Di Paolo G, Sanchez PE. Novel App knock-in mouse model shows key features of amyloid pathology and reveals profound metabolic dysregulation of microglia. Mol Neurodegener 2022; 17:41. [PMID: 35690868 PMCID: PMC9188195 DOI: 10.1186/s13024-022-00547-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic mutations underlying familial Alzheimer's disease (AD) were identified decades ago, but the field is still in search of transformative therapies for patients. While mouse models based on overexpression of mutated transgenes have yielded key insights in mechanisms of disease, those models are subject to artifacts, including random genetic integration of the transgene, ectopic expression and non-physiological protein levels. The genetic engineering of novel mouse models using knock-in approaches addresses some of those limitations. With mounting evidence of the role played by microglia in AD, high-dimensional approaches to phenotype microglia in those models are critical to refine our understanding of the immune response in the brain. METHODS We engineered a novel App knock-in mouse model (AppSAA) using homologous recombination to introduce three disease-causing coding mutations (Swedish, Arctic and Austrian) to the mouse App gene. Amyloid-β pathology, neurodegeneration, glial responses, brain metabolism and behavioral phenotypes were characterized in heterozygous and homozygous AppSAA mice at different ages in brain and/ or biofluids. Wild type littermate mice were used as experimental controls. We used in situ imaging technologies to define the whole-brain distribution of amyloid plaques and compare it to other AD mouse models and human brain pathology. To further explore the microglial response to AD relevant pathology, we isolated microglia with fibrillar Aβ content from the brain and performed transcriptomics and metabolomics analyses and in vivo brain imaging to measure energy metabolism and microglial response. Finally, we also characterized the mice in various behavioral assays. RESULTS Leveraging multi-omics approaches, we discovered profound alteration of diverse lipids and metabolites as well as an exacerbated disease-associated transcriptomic response in microglia with high intracellular Aβ content. The AppSAA knock-in mouse model recapitulates key pathological features of AD such as a progressive accumulation of parenchymal amyloid plaques and vascular amyloid deposits, altered astroglial and microglial responses and elevation of CSF markers of neurodegeneration. Those observations were associated with increased TSPO and FDG-PET brain signals and a hyperactivity phenotype as the animals aged. DISCUSSION Our findings demonstrate that fibrillar Aβ in microglia is associated with lipid dyshomeostasis consistent with lysosomal dysfunction and foam cell phenotypes as well as profound immuno-metabolic perturbations, opening new avenues to further investigate metabolic pathways at play in microglia responding to AD-relevant pathogenesis. The in-depth characterization of pathological hallmarks of AD in this novel and open-access mouse model should serve as a resource for the scientific community to investigate disease-relevant biology.
Collapse
Affiliation(s)
- Dan Xia
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Steve Lianoglou
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Thomas Sandmann
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Meredith Calvert
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Jung H. Suh
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Elliot Thomsen
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Jason Dugas
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Michelle E. Pizzo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Sarah L. DeVos
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Timothy K. Earr
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Chia-Ching Lin
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Sonnet Davis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Connie Ha
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Amy Wing-Sze Leung
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Hoang Nguyen
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Roni Chau
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Ernie Yulyaningsih
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Isabel Lopez
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Hilda Solanoy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Shababa T. Masoud
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Chun-chi Liang
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Karin Lin
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Giuseppe Astarita
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Nathalie Khoury
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Joy Yu Zuchero
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Robert G. Thorne
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
- Department of Pharmaceutics, University of Minnesota, 9-177 Weaver-Densford Hall, 308 Harvard St. SE, Minneapolis, MN 55455 USA
| | - Kevin Shen
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
- Department of Neurology, University of California, San Francisco, CA 94158 USA
| | - Stephanie Miller
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
- Department of Neurology, University of California, San Francisco, CA 94158 USA
| | - Jorge J. Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
- Department of Neurology, University of California, San Francisco, CA 94158 USA
| | | | | | | | | | - Selina Hummel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Karin Wind
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea Kunze
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Michael Willem
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig- Maximilians-Universität, München, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Daniel Barnett
- Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY USA
| | - Till S. Zimmer
- Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Anna G. Orr
- Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, New York, NY USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY USA
| | - Kimberly Scearce-Levie
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Joseph W. Lewcock
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Gilbert Di Paolo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| | - Pascal E. Sanchez
- Denali Therapeutics, Inc., 161 Oyster Point Blvd, South San Francisco, California, 94080 USA
| |
Collapse
|
77
|
Jacob MA, Peters N, Cai M, Duering M, Engelter ST, Kuhle J, de Leeuw FE, Tuladhar AM. Increased Neurofilament Light Chain Is Associated with Increased Risk of Long-Term Mortality in Cerebral Small Vessel Disease. J Stroke 2022; 24:296-299. [PMID: 35677985 PMCID: PMC9194543 DOI: 10.5853/jos.2021.04385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/08/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Mina A. Jacob
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nils Peters
- Stroke Center and Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
- University Center for Medicine of Aging, Felix Platter Hospital, Basel, Switzerland
- Stroke Center, Klinik Hirslanden, Zürich, Switzerland
| | - Mengfei Cai
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marco Duering
- Medical Image Analysis Center (MIAC AG) and Quantitative Biomedical Imaging Group (qbig), Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Stefan T. Engelter
- Stroke Center and Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
- University Center for Medicine of Aging, Felix Platter Hospital, Basel, Switzerland
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Department of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anil M. Tuladhar
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Correspondence: Anil M. Tuladhar Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Reinier Postlaan 4, PO Box 9101, 6500 HB Nijmegen, The Netherlands Tel: +31-24-361-6600 Fax: +31-24-354-1122 E-mail:
| |
Collapse
|
78
|
Vijiaratnam N, Lawton M, Heslegrave AJ, Guo T, Tan M, Jabbari E, Real R, Woodside J, Grosset K, Chelban V, Athauda D, Girges C, Barker RA, Hardy J, Wood N, Houlden H, Williams N, Ben-Shlomo Y, Zetterberg H, Grosset DG, Foltynie T, Morris HR. Combining biomarkers for prognostic modelling of Parkinson's disease. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328365. [PMID: 35577512 PMCID: PMC9279845 DOI: 10.1136/jnnp-2021-328365] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patients with Parkinson's disease (PD) have variable rates of progression. More accurate prediction of progression could improve selection for clinical trials. Although some variance in clinical progression can be predicted by age at onset and phenotype, we hypothesise that this can be further improved by blood biomarkers. OBJECTIVE To determine if blood biomarkers (serum neurofilament light (NfL) and genetic status (glucocerebrosidase, GBA and apolipoprotein E (APOE))) are useful in addition to clinical measures for prognostic modelling in PD. METHODS We evaluated the relationship between serum NfL and baseline and longitudinal clinical measures as well as patients' genetic (GBA and APOE) status. We classified patients as having a favourable or an unfavourable outcome based on a previously validated model, and explored how blood biomarkers compared with clinical variables in distinguishing prognostic phenotypes . RESULTS 291 patients were assessed in this study. Baseline serum NfL was associated with baseline cognitive status. Nfl predicted a shorter time to dementia, postural instability and death (dementia-HR 2.64; postural instability-HR 1.32; mortality-HR 1.89) whereas APOEe4 status was associated with progression to dementia (dementia-HR 3.12, 95% CI 1.63 to 6.00). NfL levels and genetic variables predicted unfavourable progression to a similar extent as clinical predictors. The combination of clinical, NfL and genetic data produced a stronger prediction of unfavourable outcomes compared with age and gender (area under the curve: 0.74-age/gender vs 0.84-ALL p=0.0103). CONCLUSIONS Clinical trials of disease-modifying therapies might usefully stratify patients using clinical, genetic and NfL status at the time of recruitment.
Collapse
Affiliation(s)
- Nirosen Vijiaratnam
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Michael Lawton
- Population Health Sciences, University of Bristol, Bristol, UK
- Department of Social Medicine, University of Bristol, Bristol, UK
| | - Amanda J Heslegrave
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Tong Guo
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Manuela Tan
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Edwin Jabbari
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - John Woodside
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Katherine Grosset
- Department of Neurology, Southern General Hospital, University of Glasgow and Institute of Neurological Sciences, Glasgow, UK
| | - Viorica Chelban
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Dilan Athauda
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Christine Girges
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Roger A Barker
- Cambridge Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - John Hardy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
- Molecular Neuroscience, University College London Institute of Neurology, London, UK
| | - Nicholas Wood
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Henry Houlden
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, UK
| | - Nigel Williams
- Cardiff University, Cardiff University Institute of Psychological Medicine and Clinical Neurosciences, Cardiff, UK
| | - Yoav Ben-Shlomo
- Department of Social Medicine, University of Bristol, Bristol, UK
| | - Henrik Zetterberg
- Dementia Research Institute, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Hong Kong Center, for Neurodegenerative Diseases, Hong Kong, People's Republic of China
| | - Donald G Grosset
- Department of Neurology, Southern General Hospital, University of Glasgow and Institute of Neurological Sciences, Glasgow, UK
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
79
|
Kang MS, Shin M, Ottoy J, Aliaga AA, Mathotaarachchi S, Quispialaya K, Pascoal TA, Collins DL, Chakravarty MM, Mathieu A, Sandelius Å, Blennow K, Zetterberg H, Massarweh G, Soucy JP, Cuello AC, Gauthier S, Waterston M, Yoganathan N, Lessard E, Haqqani A, Rennie K, Stanimirovic D, Chakravarthy B, Rosa-Neto P. Preclinical in vivo longitudinal assessment of KG207-M as a disease-modifying Alzheimer's disease therapeutic. J Cereb Blood Flow Metab 2022; 42:788-801. [PMID: 34378436 PMCID: PMC9014686 DOI: 10.1177/0271678x211035625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In vivo biomarker abnormalities provide measures to monitor therapeutic interventions targeting amyloid-β pathology as well as its effects on downstream processes associated with Alzheimer's disease pathophysiology. Here, we applied an in vivo longitudinal study design combined with imaging and cerebrospinal fluid biomarkers, mirroring those used in human clinical trials to assess the efficacy of a novel brain-penetrating anti-amyloid fusion protein treatment in the McGill-R-Thy1-APP transgenic rat model. The bi-functional fusion protein consisted of a blood-brain barrier crossing single domain antibody (FC5) fused to an amyloid-β oligomer-binding peptide (ABP) via Fc fragment of mouse IgG (FC5-mFc2a-ABP). A five-week treatment with FC5-mFc2a-ABP (loading dose of 30 mg/Kg/iv followed by 15 mg/Kg/week/iv for four weeks) substantially reduced brain amyloid-β levels as measured by positron emission tomography and increased the cerebrospinal fluid amyloid-β42/40 ratio. In addition, the 5-week treatment rectified the cerebrospinal fluid neurofilament light chain concentrations, resting-state functional connectivity, and hippocampal atrophy measured using magnetic resonance imaging. Finally, FC5-mFc2a-ABP (referred to as KG207-M) treatment did not induce amyloid-related imaging abnormalities such as microhemorrhage. Together, this study demonstrates the translational values of the designed preclinical studies for the assessment of novel therapies based on the clinical biomarkers providing tangible metrics for designing early-stage clinical trials.
Collapse
Affiliation(s)
- Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Julie Ottoy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada
| | - Arturo Aliaga Aliaga
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Kely Quispialaya
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | | | - Axel Mathieu
- Douglas Mental Health University Institute, Montreal, Canada
| | - Åsa Sandelius
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Gassan Massarweh
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | | | | | - Etienne Lessard
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Arsalan Haqqani
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Kerry Rennie
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Danica Stanimirovic
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Balu Chakravarthy
- Human Health Therapeutics, National Research Council of Canada, Ottawa, ON, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studying in Aging, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
80
|
Lin CW, Lai TT, Chen SJ, Lin CH. Elevated α-synuclein and NfL levels in tear fluids and decreased retinal microvascular densities in patients with Parkinson's disease. GeroScience 2022; 44:1551-1562. [PMID: 35484471 DOI: 10.1007/s11357-022-00576-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 12/21/2022] Open
Abstract
The pathognomonic hallmark of Parkinson's disease (PD), α-synuclein, has been observed in the retina of PD patients. We investigated whether biomarkers in the tears and retinal microvascular changes associate with PD risk and progression. This prospective study enrolled 49 PD patients and 45 age-matched healthy controls. The α-synuclein and neurofilament light chain (NfL) levels were measured using an electrochemiluminescence immunoassay. Retinal vessel density was assessed using optical coherence tomography angiography (OCT-A). The Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and Mini-Mental State Examination score were used to assess motor and cognitive progression. The α-synuclein and NfL levels in the tears were higher in PD patients than in controls (α-synuclein: 55.49 ± 8.12 pg/mL vs. 31.71 ± 3.25 pg/mL, P = 0.009; NfL: 2.89 ± 0.52 pg/mL vs. 1.47 ± 0.23 pg/mL, P = 0.02). The vessel densities in the deep plexus of central macula and the radial peripapillary capillary layer of disc region were lower in PD patients with moderate-stage compared with early-stage PD (P < 0.05). The accuracy of predicting PD occurrence using age and sex alone (area under the curve [AUC] 0.612) was significantly improved by adding α-synuclein and NfL levels and retinal vascular densities (AUC 0.752, P = 0.001). After a mean follow-up of 1.5 ± 0.3 years, the accuracy of predicting motor or cognitive progression using age, sex, and baseline motor severity as a basic model was increased by incorporating retinal microvascular and biofluid markers as a full model (P = 0.001). Our results showed that retinal microvascular densities combined with α-synuclein and NfL levels in tears are associated with risk and progression of PD.
Collapse
Affiliation(s)
- Chao-Wen Lin
- Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Ting Lai
- Department of Ophthalmology, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
81
|
Eisenstein SA, Boodram RS, Sutphen CL, Lugar HM, Gordon BA, Marshall BA, Urano F, Fagan AM, Hershey T. Plasma Neurofilament Light Chain Levels Are Elevated in Children and Young Adults With Wolfram Syndrome. Front Neurosci 2022; 16:795317. [PMID: 35495027 PMCID: PMC9039397 DOI: 10.3389/fnins.2022.795317] [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: 10/14/2021] [Accepted: 03/04/2022] [Indexed: 11/23/2022] Open
Abstract
Wolfram syndrome is a rare disease caused by pathogenic variants in the WFS1 gene with progressive neurodegeneration. As an easily accessible biomarker of progression of neurodegeneration has not yet been found, accurate tracking of the neurodegenerative process over time requires assessment by costly and time-consuming clinical measures and brain magnetic resonance imaging (MRI). A blood-based measure of neurodegeneration, neurofilament light chain (NfL), is relatively inexpensive and can be repeatedly measured at remote sites, standardized, and measured in individuals with MRI contraindications. To determine whether NfL levels may be of use in disease monitoring and reflect disease activity in Wolfram syndrome, plasma NfL levels were compared between children and young adults with Wolfram syndrome (n = 38) and controls composed of their siblings and parents (n = 35) and related to clinical severity and selected brain region volumes within the Wolfram group. NfL levels were higher in the Wolfram group [median (interquartile range) NfL = 11.3 (7.8-13.9) pg/mL] relative to controls [5.6 (4.5-7.4) pg/mL]. Within the Wolfram group, higher NfL levels related to worse visual acuity, color vision and smell identification, smaller brainstem and thalamic volumes, and faster annual rate of decrease in thalamic volume over time. Our findings suggest that plasma NfL levels can be a powerful tool to non-invasively assess underlying neurodegenerative processes in children, adolescents and young adults with Wolfram syndrome.
Collapse
Affiliation(s)
- Sarah A. Eisenstein
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Raveena S. Boodram
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Courtney L. Sutphen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Heather M. Lugar
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian A. Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Bess A. Marshall
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
- Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Fumihiko Urano
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO, United States
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Tamara Hershey
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
82
|
Ziemssen T, Arnold DL, Alvarez E, Cross AH, Willi R, Li B, Kukkaro P, Kropshofer H, Ramanathan K, Merschhemke M, Kieseier B, Su W, Häring DA, Hauser SL, Kappos L, Kuhle J. Prognostic Value of Serum Neurofilament Light Chain for Disease Activity and Worsening in Patients With Relapsing Multiple Sclerosis: Results From the Phase 3 ASCLEPIOS I and II Trials. Front Immunol 2022; 13:852563. [PMID: 35432382 PMCID: PMC9009385 DOI: 10.3389/fimmu.2022.852563] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/07/2022] [Indexed: 11/27/2022] Open
Abstract
Objective This study aims to confirm the prognostic value of baseline serum neurofilament light chain (sNfL) for on-study disease activity and worsening in patients with relapsing MS (RMS). Background Previous post-hoc studies suggested that sNfL could be a prognostic biomarker in RMS. In the phase 3 ASCLEPIOS I/II trials in which ofatumumab demonstrated better efficacy outcomes than teriflunomide, treatment with ofatumumab also led to significantly reduced sNfL levels compared to teriflunomide treatment. Design/Methods In this study, we report protocol-planned analyses from the pooled ASCLEPIOS I/II trials (N=1882). Per protocol, patients were stratified by median baseline sNfL levels (9.3 pg/ml) into high (>median) and low (≤median) categories to prognosticate: annualized rate of new/enlarging T2 (neT2) lesions in year 1 and 2, annualized relapse rate, annual percentage change in whole brain (WB) and regional brain volume [thalamus, white matter (WM), cortical gray matter (cGM)], and disability outcomes. Similar analyses were performed for the recently diagnosed (within 3 years), treatment-naive patients (no prior disease-modifying therapy) subgroup. Results High versus low sNfL at baseline was prognostic of increased on-study T2 lesion formation at year 1 (relative increase: ofatumumab +158%; teriflunomide +69%, both p<0.001), which persisted in year 2 (+65%, p=0.124; +46%, p=0.003); of higher annual percentage change of WB volume (ofatumumab, −0.32% vs. −0.24%, p=0.044, and teriflunomide, −0.43% vs. −0.29%, p=0.002), thalamic volume (−0.56% vs. −0.31%, p=0.047 and −0.94% vs. −0.49%, p<0.001), and WM volume (−0.30% vs. −0.19%, p=0.083 and −0.38% vs. −0.18%, p=0.003) but not of cGM volume (−0.39% vs. −0.32%, p=0.337 and −0.49% vs. −0.46%, p=0.563). A single sNfL assessment at baseline was not prognostic for on-study relapses or disability worsening. Results were similar in the subgroup of recently diagnosed, treatment-naive patients. Conclusion This study confirms that baseline sNfL levels are prognostic of future on-study lesion formation and whole brain and regional atrophy in all RMS patients, including recently diagnosed, treatment-naive patients.
Collapse
Affiliation(s)
- Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Clinic Carl-Gustav Carus, Dresden, Germany
- *Correspondence: Tjalf Ziemssen,
| | - Douglas L. Arnold
- Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- NeuroRx Research, Montreal, QC, Canada
| | - Enrique Alvarez
- Department of Neurology, Rocky Mountain MS Center at the University of Colorado, Aurora, CO, United States
| | - Anne H. Cross
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, United States
| | | | - Bingbing Li
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, United States
| | | | | | | | | | | | - Wendy Su
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, United States
| | | | - Stephen L. Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Ludwig Kappos
- Neurologic Clinic and Policlinic and MS Center, Department of Head, Spine and Neuromedicine, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurologic Clinic and Policlinic and MS Center, Department of Head, Spine and Neuromedicine, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
83
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
84
|
Serum NfL in Alzheimer Dementia: Results of the Prospective Dementia Registry Austria. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58030433. [PMID: 35334608 PMCID: PMC8955532 DOI: 10.3390/medicina58030433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: The neurofilament light chain (NfL) is a biomarker for neuro-axonal injury in various acute and chronic neurological disorders, including Alzheimer’s disease (AD). We here investigated the cross-sectional and longitudinal associations between baseline serum NfL (sNfL) levels and cognitive, behavioural as well as MR volumetric findings in the Prospective Dementia Registry Austria (PRODEM-Austria). Materials and Methods: All participants were clinically diagnosed with AD according to NINCDS-ADRDA criteria and underwent a detailed clinical assessment, cognitive testing (including the Mini Mental State Examination (MMSE) and the Consortium to Establish a Registry for Alzheimer’s Disease (CERAD)), the neuropsychiatric inventory (NPI) and laboratory evaluation. A total of 237 patients were included in the study. Follow-up examinations were done at 6 months, 1 year and 2 years with 93.3% of patients undergoing at least one follow-up. We quantified sNfL by a single molecule array (Simoa). In a subgroup of 125 subjects, brain imaging data (1.5 or 3T MRI, with 1 mm isotropic resolution) were available. Brain volumetry was assessed using the FreeSurfer image analysis suite (v6.0). Results: Higher sNfL concentrations were associated with worse performance in cognitive tests at baseline, including CERAD (B = −10.084, SE = 2.999, p < 0.001) and MMSE (B = −3.014, SE = 1.293, p = 0.021). The sNfL levels also correlated with the presence of neuropsychiatric symptoms (NPI total score: r = 0.138, p = 0.041) and with smaller volumes of the temporal lobe (B = −0.012, SE = 0.003, p = 0.001), the hippocampus (B = −0.001, SE = 0.000201, p = 0.013), the entorhinal (B = −0.000308, SE = 0.000124, p = 0.014), and the parahippocampal cortex (B = −0.000316, SE = 0.000113, p = 0.006). The sNfL values predicted more pronounced cognitive decline over the mean follow-up period of 22 months, but there were no significant associations with respect to change in neuropsychiatric symptoms and brain volumetric measures. Conclusions: the sNfL levels relate to cognitive, behavioural, and imaging hallmarks of AD and predicts short term cognitive decline.
Collapse
|
85
|
Rook ME, Southwell AL. Antisense Oligonucleotide Therapy: From Design to the Huntington Disease Clinic. BioDrugs 2022; 36:105-119. [PMID: 35254632 PMCID: PMC8899000 DOI: 10.1007/s40259-022-00519-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Huntington disease (HD) is a fatal progressive neurodegenerative disorder caused by an inherited mutation in the huntingtin (HTT) gene, which encodes mutant HTT protein. Though HD remains incurable, various preclinical studies have reported a favorable response to HTT suppression, emphasizing HTT lowering strategies as prospective disease-modifying treatments. Antisense oligonucleotides (ASOs) lower HTT by targeting transcripts and are well suited for treating neurodegenerative disorders as they distribute broadly throughout the central nervous system (CNS) and are freely taken up by neurons, glia, and ependymal cells. With the FDA approval of an ASO therapy for another disease of the CNS, spinal muscular atrophy, ASOs have become a particularly attractive therapeutic option for HD. However, two types of ASOs were recently assessed in human clinical trials for the treatment of HD, and both were halted early. In this review, we will explore the differences in chemistry, targeting, and specificity of these HTT ASOs as well as preliminary clinical findings and potential reasons for and implications of these halted trials.
Collapse
Affiliation(s)
- Morgan E Rook
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA.
| | - Amber L Southwell
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
86
|
Correia M, Silva I, Gabriel D, Simrén J, Carneiro A, Ribeiro S, Dória HM, Varela R, Aires A, Minta K, Antunes R, Felgueiras R, Castro P, Blenow K, Magalhães R, Zetterberg H, Maia LF. Early plasma biomarker dynamic profiles are associated with acute ischemic stroke outcomes. Eur J Neurol 2022; 29:1630-1642. [DOI: 10.1111/ene.15273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Manuel Correia
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Porto Portugal
| | - Isabel Silva
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
- i3S ‐ Instituto de Investigação e Inovação em Saúde University of Porto, Porto, Portugal and IBMC ‐ Instituto de Biologia Molecular e Celular, University of Porto Porto Portugal
| | - Denis Gabriel
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg 431 41 Mölndal Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital 431 80 Mölndal Sweden
| | - Angelo Carneiro
- Department of Neuroradiology Centro Hospitalar Universitário do Porto Porto Portugal
| | - Sara Ribeiro
- i3S ‐ Instituto de Investigação e Inovação em Saúde University of Porto, Porto, Portugal and Ipatimup ‐ Institute of Molecular Pathology and Immunology, University of Porto Porto Portugal
| | - Hugo Mota Dória
- Department of Neuroradiology Centro Hospitalar Universitário do Porto Porto Portugal
| | - Ricardo Varela
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
| | - Ana Aires
- Department of Neurology Centro Hospitalar Universitário de São João Porto Portugal
| | - Karolina Minta
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg 431 41 Mölndal Sweden
- Department of Neurodegenerative Disease University College London Institute of Neurology Queen Square London UK
| | - Rui Antunes
- Intensive Care Unit Centro Hospitalar Universitário do Porto Porto Portugal
| | - Rui Felgueiras
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
| | - Pedro Castro
- Department of Neurology Centro Hospitalar Universitário de São João Porto Portugal
| | - Kaj Blenow
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg 431 41 Mölndal Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital 431 80 Mölndal Sweden
| | - Rui Magalhães
- Population Studies Instituto de Ciências Biomédicas Abel Salazar University of Porto Porto Portugal
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg 431 41 Mölndal Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital 431 80 Mölndal Sweden
- Department of Neurodegenerative Disease University College London Institute of Neurology Queen Square London UK
- UK Dementia Research Institute at UCL London UK
- Hong Kong Center for Neurodegenerative Diseases Hong Kong China
| | - Luis F Maia
- Department of Neurology Centro Hospitalar Universitário do Porto Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Porto Portugal
- i3S ‐ Instituto de Investigação e Inovação em Saúde University of Porto, Porto, Portugal and IBMC ‐ Instituto de Biologia Molecular e Celular, University of Porto Porto Portugal
| |
Collapse
|
87
|
Park JE, Gunasekaran TI, Cho YH, Choi SM, Song MK, Cho SH, Kim J, Song HC, Choi KY, Lee JJ, Park ZY, Song WK, Jeong HS, Lee KH, Lee JS, Kim BC. Diagnostic Blood Biomarkers in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10010169. [PMID: 35052848 PMCID: PMC8773964 DOI: 10.3390/biomedicines10010169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Potential biomarkers for Alzheimer’s disease (AD) include amyloid β1–42 (Aβ1–42), t-Tau, p-Tau181, neurofilament light chain (NFL), and neuroimaging biomarkers. Their combined use is useful for diagnosing and monitoring the progress of AD. Therefore, further development of a combination of these biomarkers is essential. We investigated whether plasma NFL/Aβ1–42 can serve as a plasma-based primary screening biomarker reflecting brain neurodegeneration and amyloid pathology in AD for monitoring disease progression and early diagnosis. We measured the NFL and Aβ1–42 concentrations in the CSF and plasma samples and performed correlation analysis to evaluate the utility of these biomarkers in the early diagnosis and monitoring of AD spectrum disease progression. Pearson’s correlation analysis was used to analyse the associations between the fluid biomarkers and neuroimaging data. The study included 136 participants, classified into five groups: 28 cognitively normal individuals, 23 patients with preclinical AD, 22 amyloid-negative patients with amnestic mild cognitive impairment, 32 patients with prodromal AD, and 31 patients with AD dementia. With disease progression, the NFL concentrations increased and Aβ1–42 concentrations decreased. The plasma and CSF NFL/Aβ1–42 were strongly correlated (r = 0.558). Plasma NFL/Aβ1–42 was strongly correlated with hippocampal volume/intracranial volume (r = 0.409). In early AD, plasma NFL/Aβ1–42 was associated with higher diagnostic accuracy than the individual biomarkers. Moreover, in preclinical AD, plasma NFL/Aβ1–42 changed more rapidly than the CSF t-Tau or p-Tau181 concentrations. Our findings highlight the utility of plasma NFL/Aβ1–42 as a non-invasive plasma-based biomarker for early diagnosis and monitoring of AD spectrum disease progression.
Collapse
Affiliation(s)
- Jung Eun Park
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea; (J.E.P.); (T.I.G.); (Y.H.C.); (K.H.L.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Tamil Iniyan Gunasekaran
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea; (J.E.P.); (T.I.G.); (Y.H.C.); (K.H.L.)
- Gwangju Alzheimer’s Disease and Related Dementias Cohort Center, Chosun University, Gwangju 61452, Korea; (K.Y.C.); (J.J.L.)
| | - Yeong Hee Cho
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea; (J.E.P.); (T.I.G.); (Y.H.C.); (K.H.L.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
| | - Seong-Min Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Min-Kyung Song
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Jahae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (J.K.); (H.-C.S.)
| | - Ho-Chun Song
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea; (J.K.); (H.-C.S.)
| | - Kyu Yeong Choi
- Gwangju Alzheimer’s Disease and Related Dementias Cohort Center, Chosun University, Gwangju 61452, Korea; (K.Y.C.); (J.J.L.)
| | - Jang Jae Lee
- Gwangju Alzheimer’s Disease and Related Dementias Cohort Center, Chosun University, Gwangju 61452, Korea; (K.Y.C.); (J.J.L.)
| | - Zee-Yong Park
- Laboratory of Functional and Medicinal Proteomics, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Woo Keun Song
- Cell Logistics and Silver Health Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea;
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Kun Ho Lee
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea; (J.E.P.); (T.I.G.); (Y.H.C.); (K.H.L.)
- Gwangju Alzheimer’s Disease and Related Dementias Cohort Center, Chosun University, Gwangju 61452, Korea; (K.Y.C.); (J.J.L.)
- Aging Neuroscience Research Group, Korea Brain Research Institute, Daegu 41062, Korea
| | - Jung Sup Lee
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea; (J.E.P.); (T.I.G.); (Y.H.C.); (K.H.L.)
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju 61452, Korea
- Correspondence: (J.S.L.); (B.C.K.); Tel.: +82-62-220-6665 (J.S.L.); +82-62-220-6123 (B.C.K.)
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju 61469, Korea; (S.-M.C.); (S.H.C.)
- Department of Neurology, Chonnam National University Hospital, Gwangju 61469, Korea;
- Correspondence: (J.S.L.); (B.C.K.); Tel.: +82-62-220-6665 (J.S.L.); +82-62-220-6123 (B.C.K.)
| |
Collapse
|
88
|
Katz JS, Rothstein JD, Cudkowicz ME, Genge A, Oskarsson B, Hains AB, Chen C, Galanter J, Burgess BL, Cho W, Kerchner GA, Yeh FL, Ghosh AS, Cheeti S, Brooks L, Honigberg L, Couch JA, Rothenberg ME, Brunstein F, Sharma KR, van den Berg L, Berry JD, Glass JD. A Phase 1 study of GDC-0134, a dual leucine zipper kinase inhibitor, in ALS. Ann Clin Transl Neurol 2022; 9:50-66. [PMID: 35014217 PMCID: PMC8791798 DOI: 10.1002/acn3.51491] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Objective Dual leucine zipper kinase (DLK), which regulates the c‐Jun N‐terminal kinase pathway involved in axon degeneration and apoptosis following neuronal injury, is a potential therapeutic target in amyotrophic lateral sclerosis (ALS). This first‐in‐human study investigated safety, tolerability, and pharmacokinetics (PK) of oral GDC‐0134, a small‐molecule DLK inhibitor. Plasma neurofilament light chain (NFL) levels were explored in GDC‐0134‐treated ALS patients and DLK conditional knockout (cKO) mice. Methods The study included placebo‐controlled, single and multiple ascending‐dose (SAD; MAD) stages, and an open‐label safety expansion (OLE) with adaptive dosing for up to 48 weeks. Results Forty‐nine patients were enrolled. GDC‐0134 (up to 1200 mg daily) was well tolerated in the SAD and MAD stages, with no serious adverse events (SAEs). In the OLE, three study drug‐related SAEs occurred: thrombocytopenia, dysesthesia (both Grade 3), and optic ischemic neuropathy (Grade 4); Grade ≤2 sensory neurological AEs led to dose reductions/discontinuations. GDC‐0134 exposure was dose‐proportional (median half‐life = 84 h). Patients showed GDC‐0134 exposure‐dependent plasma NFL elevations; DLK cKO mice also exhibited plasma NFL compared to wild‐type littermates. Interpretation This trial characterized GDC‐0134 safety and PK, but no adequately tolerated dose was identified. NFL elevations in GDC‐0134‐treated patients and DLK cKO mice raised questions about interpretation of biomarkers affected by both disease and on‐target drug effects. The safety profile of GDC‐0134 was considered unacceptable and led to discontinuation of further drug development for ALS. Further work is necessary to understand relationships between neuroprotective and potentially therapeutic effects of DLK knockout/inhibition and NFL changes in patients with ALS.
Collapse
Affiliation(s)
- Jonathan S Katz
- Forbes Norris MDA/ALS Research Center, California Pacific Medical Center, San Francisco, California, USA
| | | | - Merit E Cudkowicz
- Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Angela Genge
- Montreal Neurological Institute & Hospital, Montreal, QC, Canada
| | | | - Avis B Hains
- Genentech, Inc., South San Francisco, California, USA
| | - Chen Chen
- Genentech, Inc., South San Francisco, California, USA
| | | | | | - William Cho
- Genentech, Inc., South San Francisco, California, USA
| | | | - Felix L Yeh
- Genentech, Inc., South San Francisco, California, USA
| | | | | | - Logan Brooks
- Genentech, Inc., South San Francisco, California, USA
| | - Lee Honigberg
- Genentech, Inc., South San Francisco, California, USA
| | | | | | | | | | | | - James D Berry
- Neurological Clinical Research Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
89
|
Dodiya HB, Lutz HL, Weigle IQ, Patel P, Michalkiewicz J, Roman-Santiago CJ, Zhang CM, Liang Y, Srinath A, Zhang X, Xia J, Olszewski M, Zhang X, Schipma MJ, Chang EB, Tanzi RE, Gilbert JA, Sisodia SS. Gut microbiota-driven brain Aβ amyloidosis in mice requires microglia. J Exp Med 2022; 219:e20200895. [PMID: 34854884 PMCID: PMC8647415 DOI: 10.1084/jem.20200895] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/16/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
We previously demonstrated that lifelong antibiotic (ABX) perturbations of the gut microbiome in male APPPS1-21 mice lead to reductions in amyloid β (Aβ) plaque pathology and altered phenotypes of plaque-associated microglia. Here, we show that a short, 7-d treatment of preweaned male mice with high-dose ABX is associated with reductions of Aβ amyloidosis, plaque-localized microglia morphologies, and Aβ-associated degenerative changes at 9 wk of age in male mice only. More importantly, fecal microbiota transplantation (FMT) from transgenic (Tg) or WT male donors into ABX-treated male mice completely restored Aβ amyloidosis, plaque-localized microglia morphologies, and Aβ-associated degenerative changes. Transcriptomic studies revealed significant differences between vehicle versus ABX-treated male mice and FMT from Tg mice into ABX-treated mice largely restored the transcriptome profiles to that of the Tg donor animals. Finally, colony-stimulating factor 1 receptor (CSF1R) inhibitor-mediated depletion of microglia in ABX-treated male mice failed to reduce cerebral Aβ amyloidosis. Thus, microglia play a critical role in driving gut microbiome-mediated alterations of cerebral Aβ deposition.
Collapse
Affiliation(s)
- Hemraj B. Dodiya
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Holly L. Lutz
- Department of Pediatrics and Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA
| | - Ian Q. Weigle
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Priyam Patel
- Center for Genetic Medicine, Northwestern University, Chicago, IL
| | | | | | | | - Yingxia Liang
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Abhinav Srinath
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Xulun Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Jessica Xia
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Monica Olszewski
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | - Xiaoqiong Zhang
- Department of Neurobiology, The University of Chicago, Chicago, IL
| | | | - Eugene B. Chang
- Department of Digestive Diseases, The University of Chicago, Chicago, IL
| | | | - Jack A. Gilbert
- Department of Pediatrics and Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA
| | | |
Collapse
|
90
|
Clinical Application of Plasma Neurofilament Light Chain in a Memory Clinic: A Pilot Study. Dement Neurocogn Disord 2022; 21:59-70. [PMID: 35585907 PMCID: PMC9085534 DOI: 10.12779/dnd.2022.21.2.59] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose Neurofilament light chain (NfL) has been considered as a biomarker for neurodegenerative diseases including Alzheimer’s disease (AD). We measured plasma NfL levels in older adults with cognitive complaints and evaluated their clinical usefulness in AD. Methods Plasma levels of NfL, measured by using the single molecule array method, were acquired in a total of 113 subjects consisting of subjective cognitive decline (SCD; n=14), mild cognitive impairment (MCI; n=37), or dementia of Alzheimer type (DAT; n=62). Plasma NfL level was compared among three groups, and its association with cognitive and functional status was also analyzed. Results After adjusting for age, plasma NfL level was higher in subjects with DAT (65.98±84.96 pg/mL), compared to in subjects with SCD (16.90±2.54 pg/mL) or MCI (25.53±10.42 pg/mL, p=0.004). NfL levels were correlated with scores of the mini-mental state examination (r=−0.242, p=0.021), clinical dementia rating (CDR) (r=0.291, p=0.005), or CDR-sum of boxes (r=0.276, p=0.008). Just for participants who performed amyloid positron emission tomography (PET), the levels were different between subjects with PET (−) (n=17, 25.95±13.25 pg/mL) and PET (+) (n=16, 63.65±81.90 pg/mL, p=0.010). Additionally, plasma NfL levels were different between vascular dementia and vascular MCI, and between Parkinson’s disease- dementia and no dementia. Conclusions This pilot study shows that in subjects with DAT, plasma NfL levels increase. Plasma NfL level correlated with cognitive and functional status. Further longitudinal studies may help to apply the plasma NfL levels to AD, as a potential biomarker for the diagnosis and predicting progression.
Collapse
|
91
|
Agnello L, Lo Sasso B, Vidali M, Scazzone C, Piccoli T, Gambino CM, Bivona G, Giglio RV, Ciaccio AM, La Bella V, Ciaccio M. Neurogranin as a Reliable Biomarker for Synaptic Dysfunction in Alzheimer's Disease. Diagnostics (Basel) 2021; 11:diagnostics11122339. [PMID: 34943576 PMCID: PMC8700711 DOI: 10.3390/diagnostics11122339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Neurogranin is a post-synaptic protein expressed in the neurons of the hippocampus and cerebral cortex. It has been recently proposed as a promising biomarker of synaptic dysfunction, especially in Alzheimer's disease (AD). However, more efforts are needed before introducing it in clinical practice, including the definition of its reference interval (RI). The aim of the study was to establish the RI of cerebrospinal fluid (CSF) neurogranin levels in controls and individuals with non-neurodegenerative neurological diseases; (2) We included a total of 136 individuals that were sub-grouped as follows: AD patients (n = 33), patients with non-neurodegenerative neurological diseases (n = 70) and controls (33). We measured CSF neurogranin levels by a commercial ELISA kit. CSF RI of neurogranin was calculated by a robust method; (3) Results: AD patients showed increased levels of neurogranin. We also found that neurogranin was significantly correlated with T-tau, P-tau and mini mental state examination in AD patients. The lower and upper reference limits of the RI were 2.9 (90%CI 0.1-10.8) and 679 (90%CI 595-779), respectively; (4) Conclusion: This is the first study establishing the RI of CSF neurogranin.
Collapse
Affiliation(s)
- Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
- Department of Laboratory Medicine, Azienda Ospedaliera Universitaria Policlinico “P. Giaccone”, 90127 Palermo, Italy
| | - Matteo Vidali
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Concetta Scazzone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Tommaso Piccoli
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| | - Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Giulia Bivona
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
| | - Anna Maria Ciaccio
- Unit of Clinical Biochemistry, University of Palermo, 90127 Palermo, Italy;
| | - Vincenzo La Bella
- ALS Clinical Research Center, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy;
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (L.A.); (B.L.S.); (C.S.); (C.M.G.); (G.B.); (R.V.G.)
- Department of Laboratory Medicine, Azienda Ospedaliera Universitaria Policlinico “P. Giaccone”, 90127 Palermo, Italy
- Correspondence: ; Tel.: +39-0916553296
| |
Collapse
|
92
|
Sandebring-Matton A, Axenhus M, Bogdanovic N, Winblad B, Schedin-Weiss S, Nilsson P, Tjernberg LO. Microdissected Pyramidal Cell Proteomics of Alzheimer Brain Reveals Alterations in Creatine Kinase B-Type, 14-3-3-γ, and Heat Shock Cognate 71. Front Aging Neurosci 2021; 13:735334. [PMID: 34867272 PMCID: PMC8641652 DOI: 10.3389/fnagi.2021.735334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
Novel insights on proteins involved in Alzheimer’s disease (AD) are needed. Since multiple cell types and matrix components are altered in AD, bulk analysis of brain tissue maybe difficult to interpret. In the current study, we isolated pyramidal cells from the cornu ammonis 1 (CA1) region of the hippocampus from five AD and five neurologically healthy donors using laser capture microdissection (LCM). The samples were analyzed by proteomics using 18O-labeled internal standard and nano-high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) for relative quantification. Fold change between AD and control was calculated for the proteins that were identified in at least two individual proteomes from each group. From the 10 cases analyzed, 62 proteins were identified in at least two AD cases and two control cases. Creatine kinase B-type (CKB), 14-3-3-γ, and heat shock cognate 71 (Hsc71), which have not been extensively studied in the context of the human AD brain previously, were selected for further studies by immunohistochemistry (IHC). In hippocampus, semi-quantitative measures of IHC staining of the three proteins confirmed the findings from our proteomic analysis. Studies of the same proteins in the frontal cortex revealed that the alterations remained for CKB and 14-3-3-γ but not for Hsc71. Protein upregulation in CA1 neurons of final stage AD is either a result of detrimental, pathological effects, or from cell-specific protective response mechanisms in surviving neurons. Based on previous findings from experimental studies, CKB and Hsc71 likely exhibit protective effects, whereas 14-3-3-γ may represent a detrimental pathway. These new players could reflect pathways of importance for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Anna Sandebring-Matton
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Michael Axenhus
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Nenad Bogdanovic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Clinical Chemistry, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
93
|
Diekämper E, Brix B, Stöcker W, Vielhaber S, Galazky I, Kreissl MC, Genseke P, Düzel E, Körtvelyessy P. Neurofilament Levels Are Reflecting the Loss of Presynaptic Dopamine Receptors in Movement Disorders. Front Neurosci 2021; 15:690013. [PMID: 34924923 PMCID: PMC8681873 DOI: 10.3389/fnins.2021.690013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 10/07/2021] [Indexed: 12/01/2022] Open
Abstract
Aims: Neurofilament light chain (NfL) and phosphorylated neurofilament heavy chain (pNfH) are biomarkers for neuroaxonal damage. We assessed whether NfL and other biomarker levels in the CSF are correlated to the loss of presynaptic dopamine transporters in neurons as detected with dopamine transporter SPECT (DaTscan). Methods: We retrospectively identified 47 patients (17 Alzheimer's dementia, 10 idiopathic Parkinson's disease, 7 Lewy body dementia, 13 progressive supranuclear palsy or corticobasal degeneration) who received a DaTscan and a lumbar puncture. DaTscan imaging was performed according to current guidelines, and z-scores indicating the decrease in uptake were software based calculated for the nucleus caudatus and putamen. The CSF biomarkers progranulin, total-tau, alpha-synuclein, NfL, and pNfH were correlated with the z-scores. Results: DaTscan results in AD patients did not correlate with any biomarker. Subsuming every movement disorder with nigrostriatal neurodegeneration resulted in a strong correlation between putamen/nucleus caudatus and NfL (nucleus caudatus right p < 0.01, putamen right p < 0.05, left p < 0.05) and between pNfH and putamen (right p < 0.05; left p < 0.042). Subdividing in disease cohorts did not reveal significant correlations. Progranulin, alpha-synuclein, and total-tau did not correlate with DaTscan results. Conclusion: We show a strong correlation of NfL and pNfH with pathological changes in presynaptic dopamine transporter density in the putamen concomitant to nigrostriatal degeneration. This correlation might explain the reported correlation of impaired motor functions in PD and NfL as seen before, despite the pathological heterogeneity of these diseases.
Collapse
Affiliation(s)
- Elena Diekämper
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Britta Brix
- Institute for Experimental Immunology, EUROIMMUN Medizinische Labordiagnostika AG, Lübeck, Germany
| | - Winfried Stöcker
- Clinical-Immunological Laboratory Prof. Dr. Stöcker, Lübeck, Germany
| | - Stefan Vielhaber
- Department of Neurology, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Imke Galazky
- Department of Neurology, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Michael C. Kreissl
- Department of Nuclear Medicine, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Philipp Genseke
- Department of Nuclear Medicine, University Hospital Magdeburg, Otto-von Guericke University, Magdeburg, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute for Cognitive Neurology and Dementia Research, Magdeburg, Germany
| | - Péter Körtvelyessy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Neuropathology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
94
|
De Lorenzo R, Loré NI, Finardi A, Mandelli A, Cirillo DM, Tresoldi C, Benedetti F, Ciceri F, Rovere-Querini P, Comi G, Filippi M, Manfredi AA, Furlan R. Blood neurofilament light chain and total tau levels at admission predict death in COVID-19 patients. J Neurol 2021; 268:4436-4442. [PMID: 33973106 PMCID: PMC8108733 DOI: 10.1007/s00415-021-10595-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Patients infected with SARS-CoV-2 range from asymptomatic, to mild, moderate or severe disease evolution including fatal outcome. Thus, early predictors of clinical outcome are highly needed. We investigated markers of neural tissue damage as a possible early sign of multisystem involvement to assess their clinical prognostic value on survival or transfer to intensive care unit (ICU). METHODS We collected blood from 104 patients infected with SARS-CoV-2 the day of admission to the emergency room and measured blood neurofilament light chair (NfL), glial fibrillary acidic protein (GFAP), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), and total tau protein levels. RESULTS We found that NfL, GFAP, and tau were significantly increased in patients with fatal outcome, while NfL and UCH-L1 in those needing ICU transfer. ROC and Kaplan-Meier curves indicated that total tau levels at admission accurately predict mortality. CONCLUSIONS Blood neural markers may provide additional prognostic value to conventional biomarkers used to predict COVID-19 outcome.
Collapse
Affiliation(s)
- Rebecca De Lorenzo
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Nicola I Loré
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Annamaria Finardi
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Mandelli
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristina Tresoldi
- Hematology and Bone Marrow Transplant, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, Milan, Italy
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giancarlo Comi
- Vita-Salute San Raffaele University, Milan, Italy
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Filippi
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, Neurology Unit, Neurophysiology Service, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angelo A Manfredi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Roberto Furlan
- Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
95
|
Yue X, Zhou Y, Qiao M, Zhao X, Huang X, Zhao T, Cheng X, Fan M, Zhao Y, Chen R, Zhu L. Intermittent hypoxia treatment alleviates memory impairment in the 6-month-old APPswe/PS1dE9 mice and reduces amyloid beta accumulation and inflammation in the brain. Alzheimers Res Ther 2021; 13:194. [PMID: 34844651 PMCID: PMC8630860 DOI: 10.1186/s13195-021-00935-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Background Alzheimer’s disease (AD) is a progressive, degenerative, and terminal disease without cure. There is an urgent need for a new strategy to treat AD. The aim of this study was to investigate the effects of intermittent hypoxic treatment (IHT) on cognitive functions in a mouse model of AD and unravel the mechanism of action of IHT. Methods Six-month-old APPswe/PS1dE9 (APP/PS1) male mice were exposed to hypoxic environment (14.3% O2) 4 h/day for 14 days or 28 days. Cognitive functions were measured by Morris water maze test after either 14 days or 42 days of interval. Thereafter the distribution of amyloid plaque and microglial activation were determined by mouse brain immunohistochemistry, while the amyloid beta (Aβ) and inflammatory cytokines were measured by ELISA and Western Blot. Microarray was used for studying gene expressions in the hippocampus. Results IHT for 14 days or 28 days significantly improved the spatial memory ability of the 6-month-old APP/PS1 mice. The memory improvement by 14 days IHT lasted to 14 days, but not to 42 days. The level of Aβ plaques and neurofilament accumulations was reduced markedly after the IHT exposure. IHT reduced the pro-inflammatory cytokines IL-1β, IL-6 levels, and β-secretase cleavage of APP processing which implies reduced Aβ production. Microarray analysis revealed a large number of genes in the hippocampus were significantly altered which are known to be metabolism-regulated genes. Conclusions This study provides evidence of the beneficial effect of IHT on the progression of AD by alleviating memory impairment, reducing Aβ accumulation and inflammation in the brain. IHT can be developed as a novel measure to relieve the progression of AD by targeting multiple pathways in the AD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00935-z.
Collapse
Affiliation(s)
- Xiangpei Yue
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yanzhao Zhou
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Meng Qiao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xingnan Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Tong Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xiang Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ming Fan
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yongqi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ruoli Chen
- Institute for Science and Technology in Medicine, School of Pharmacy, Keele University, Kelle, UK.
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China. .,Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China. .,Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
96
|
Laudanski K, Hajj J, Restrepo M, Siddiq K, Okeke T, Rader DJ. Dynamic Changes in Central and Peripheral Neuro-Injury vs. Neuroprotective Serum Markers in COVID-19 Are Modulated by Different Types of Anti-Viral Treatments but Do Not Affect the Incidence of Late and Early Strokes. Biomedicines 2021; 9:1791. [PMID: 34944606 PMCID: PMC8698659 DOI: 10.3390/biomedicines9121791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/07/2023] Open
Abstract
The balance between neurodegeneration, neuroinflammation, neuroprotection, and COVID-19-directed therapy may underly the heterogeneity of SARS-CoV-2's neurological outcomes. A total of 105 patients hospitalized with a diagnosis of COVID-19 had serum collected over a 6 month period to assess neuroinflammatory (MIF, CCL23, MCP-1), neuro-injury (NFL, NCAM-1), neurodegenerative (KLK6, τ, phospho τ, amyloids, TDP43, YKL40), and neuroprotective (clusterin, fetuin, TREM-2) proteins. These were compared to markers of nonspecific inflammatory responses (IL-6, D-dimer, CRP) and of the overall viral burden (spike protein). Data regarding treatment (steroids, convalescent plasma, remdasavir), pre-existing conditions, and incidences of strokes were collected. Amyloid β42, TDP43, NF-L, and KLK6 serum levels declined 2-3 days post-admission, yet recovered to admission baseline levels by 7 days. YKL-40 and NCAM-1 levels remained elevated over time, with clusters of differential responses identified among TREM-2, TDP43, and YKL40. Fetuin was elevated after the onset of COVID-19 while TREM-2 initially declined before significantly increasing over time. MIF serum level was increased 3-7 days after admission. Ferritin correlated with TDP-43 and KLK6. No treatment with remdesivir coincided with elevations in Amyloid-β40. A lack of convalescent plasma resulted in increased NCAM-1 and total tau, and steroidal treatments did not significantly affect any markers. A total of 11 incidences of stroke were registered up to six months after initial admission for COVID-19. Elevated D-dimer, platelet counts, IL-6, and leukopenia were observed. Variable MIF serum levels differentiated patients with CVA from those who did not have a stroke during the acute phase of COVID-19. This study demonstrated concomitant and opposite changes in neurodegenerative and neuroprotective markers persisting well into recovery.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- The Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jihane Hajj
- School of Nursing, Widener University, Philadelphia, PA 19013, USA;
| | - Mariana Restrepo
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Kumal Siddiq
- College of Arts and Sciences, Drexel University, Philadelphia, PA 19104, USA;
| | - Tony Okeke
- School of Biomedical Engineering, Drexel University, Philadelphia, PA 19104, USA;
| | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
97
|
Luo W, Chen Y, Mao S, Jin J, Liu C, Zhong X, Sun X, Kermode AG, Qiu W. Serum neurofilament light chain in adult and pediatric patients with myelin oligodendrocyte glycoprotein antibody-associated disease: Correlation with relapses and seizures. J Neurochem 2021; 160:568-577. [PMID: 34839538 DOI: 10.1111/jnc.15549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022]
Abstract
Myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) causes major disability as a consequence of recurrent demyelinating events and neuronal loss. Biomarkers identifying different phenotypes of recurrence or tissue damage might be useful to guide individualized therapy. Herein, we evaluated serum neurofilament light chain (sNfL) as a potential biomarker in both adult and pediatric MOGAD patients. Forty-nine patients with MOGAD (37 adults, 12 children) and 71 healthy controls (HCs) (56 adults, 15 children) were enrolled prospectively from September 2019 to April 2021 at the Third Affiliated Hospital of Sun Yat-sen University and the Children's Hospital, Zhejiang University School of Medicine. sNfL levels were determined using ultrasensitive single-molecule array assay and correlated with clinical parameters. The sNfL levels in MOGAD adults in a relapsed state (median: 31.0 pg/ml) were higher than those in a remission state (8.1 pg/ml, p = 0.001) and in HC adults (10.3 pg/ml, p = 0.004). Similar results were observed in children (relapse: 46.8 pg/ml vs. remission: 13.1 pg/ml, p = 0.001; and vs. HCs: 8.2 pg/ml, p = 0.007) sNfL levels were correlated with recent relapses within 60 days (multivariate: β = 2.02, p = 0.003), seizures (multivariate: β = 2.50, p = 0.021) and brain lesions on magnetic resonance imaging (MRI) of a recent relapse (multivariate: β = 1.72, p = 0.012). Our study showed that sNfL levels are beneficial for identifying recent relapses and seizures and suggest that adult and pediatric MOGAD patients had similar sNfL levels.
Collapse
Affiliation(s)
- Wenjing Luo
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yashuang Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shanshan Mao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianing Jin
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunxin Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaonan Zhong
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaobo Sun
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Perth, Western Australia, Australia.,Department of Neurology, Sir Charles Gairdner Hospital, Queen Elizabeth II Medical Centre, Perth, Western Australia, Australia
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
98
|
Karoly HC, Skrzynski CJ, Moe EN, Bryan AD, Hutchison KE. Exploring relationships between alcohol consumption, inflammation, and brain structure in a heavy drinking sample. Alcohol Clin Exp Res 2021; 45:2256-2270. [PMID: 34523725 PMCID: PMC8642310 DOI: 10.1111/acer.14712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/09/2021] [Accepted: 09/07/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chronic alcohol consumption is associated with structural brain changes and increased inflammatory signaling throughout the brain and body. Increased inflammation in the brain has been associated with structural damage. Recent studies have also shown that neurofilament light polypeptide (NfL) is released into the systemic circulation following neuronal damage. Although NfL has thus been proposed as a biomarker for neurodegenerative diseases, its connection to alcohol use disorder has not been explored. For this secondary data analysis, we proposed a conceptual model linking alcohol consumption, the pro-inflammatory cytokine IL-6, brain structure, and NfL in heavy drinking participants. METHODS Of the 182 individuals enrolled in this study, 81 participants had usable data on gray matter (GM) thickness and 80 had usable data on white matter (WM) diffusivity. A subset of participants had NfL (n = 78) and IL-6 (n = 117) data. An estimate of GM thickness was extracted from middle frontal brain regions using FreeSurfer. Estimated mean WM diffusivity values were extracted from Tract Based Spatial Statistics. NfL and IL-6 were measured in blood. Regression models were used to test individual linkages in the conceptual model. Based on significant regression results, we created a simplified conceptual model, which we tested using path analysis. RESULTS In regressions, negative relationships emerged between GM and both drinks per drinking day (DPDD) (p = 0.018) and NfL (p = 0.004). A positive relationship emerged between WM diffusivity and DPDD (p = 0.033). IL-6 was not significantly associated with alcohol use, GM or WM. The final path model demonstrated adequate fit to the data and showed significant, negative associations between DPDD and middle frontal gyrus (MFG) thickness, and between MFG thickness and NfL, but the association between DPDD and NfL was not significant. CONCLUSIONS This is the first study to show that heavy drinking is associated with lower GM thickness and higher WM diffusivity and that lower GM thickness is associated with higher circulating NfL. The analyses also show that the effects of drinking do not involve the pro-inflammatory cytokine IL-6.
Collapse
Affiliation(s)
- Hollis C Karoly
- Institute for Cognitive Science, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Psychology, Colorado State University, Fort Collins, Colorado, USA
| | - Carillon J Skrzynski
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Erin N Moe
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Angela D Bryan
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Kent E Hutchison
- Institute for Cognitive Science, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
99
|
Guedes VA, Lai C, Devoto C, Edwards KA, Mithani S, Sass D, Vorn R, Qu BX, Rusch HL, Martin CA, Walker WC, Wilde EA, Diaz-Arrastia R, Gill JM, Kenney K. Extracellular Vesicle Proteins and MicroRNAs Are Linked to Chronic Post-Traumatic Stress Disorder Symptoms in Service Members and Veterans With Mild Traumatic Brain Injury. Front Pharmacol 2021; 12:745348. [PMID: 34690777 PMCID: PMC8526745 DOI: 10.3389/fphar.2021.745348] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/06/2021] [Indexed: 01/20/2023] Open
Abstract
Symptoms of post-traumatic stress disorder (PTSD) are common in military populations, and frequently associated with a history of combat-related mild traumatic brain injury (mTBI). In this study, we examined relationships between severity of PTSD symptoms and levels of extracellular vesicle (EV) proteins and miRNAs measured in the peripheral blood in a cohort of military service members and Veterans (SMs/Vs) with chronic mTBI(s). Participants (n = 144) were divided into groups according to mTBI history and severity of PTSD symptoms on the PTSD Checklist for DSM-5 (PCL-5). We analyzed EV levels of 798 miRNAs (miRNAs) as well as EV and plasma levels of neurofilament light chain (NfL), Tau, Amyloid beta (Aβ) 42, Aβ40, interleukin (IL)-10, IL-6, tumor necrosis factor-alpha (TNFα), and vascular endothelial growth factor (VEGF). We observed that EV levels of neurofilament light chain (NfL) were elevated in participants with more severe PTSD symptoms (PCL-5 ≥ 38) and positive mTBI history, when compared to TBI negative controls (p = 0.024) and mTBI participants with less severe PTSD symptoms (p = 0.006). Levels of EV NfL, plasma NfL, and hsa-miR-139–5p were linked to PCL-5 scores in regression models. Our results suggest that levels of NfL, a marker of axonal damage, are associated with PTSD symptom severity in participants with remote mTBI. Specific miRNAs previously linked to neurodegenerative and inflammatory processes, and glucocorticoid receptor signaling pathways, among others, were also associated with the severity of PTSD symptoms. Our findings provide insights into possible signaling pathways linked to the development of persistent PTSD symptoms after TBI and biological mechanisms underlying susceptibility to PTSD.
Collapse
Affiliation(s)
- Vivian A Guedes
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Chen Lai
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Christina Devoto
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, MD, United States
| | - Katie A Edwards
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Sara Mithani
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Dilorom Sass
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Rany Vorn
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Bao-Xi Qu
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,Center for Neuroscience and Rehabilitation Medicine, Uniformed Services University of the Health Sciences and National Institutes of Health, Bethesda, MD, United States
| | - Heather L Rusch
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Carina A Martin
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - William C Walker
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University, Richmond, VA, United States
| | - Elisabeth A Wilde
- CENC Neuroimaging Core, George E. Wahlen VA Salt Lake City Healthcare System and Traumatic Brain Injury and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jessica M Gill
- CENC Biorepository, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Johns Hopkins University School of Nursing and Medicine, Baltimore, MD, United States
| | - Kimbra Kenney
- CENC Biorepository, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
100
|
Kouchaki E, Dashti F, Mirazimi SMA, Alirezaei Z, Jafari SH, Hamblin MR, Mirzaei H. Neurofilament light chain as a biomarker for diagnosis of multiple sclerosis. EXCLI JOURNAL 2021; 20:1308-1325. [PMID: 34602928 PMCID: PMC8481790 DOI: 10.17179/excli2021-3973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
The treatments for multiple sclerosis (MS) have improved over the past 25 years, but now the main question for physicians is deciding who should receive treatment, for how long, and when to switch to other options. These decisions are typically based on treatment tolerance and a reasonable expectation of long-term efficacy. A significant unmet need is the lack of accurate laboratory measurements for diagnosis, and monitoring of treatment response, including deterioration and disease progression. There are few validated biomarkers for MS, and in practice, physicians employ two biomarkers discovered fifty years ago for MS diagnosis, often in combination with MRI scans. These biomarkers are intrathecal IgG and oligoclonal bands in the CSF (cerebrospinal fluid). Neurofilament light chain (NfL) is a relatively new biomarker for MS diagnosis and follow up. Neurofilaments are neuron-specific cytoskeleton proteins that can be measured in various body compartments. NfL is a new biomarker for MS that can be measured in serum samples, but this still needs further study to specify the laboratory cut-off values in clinical practice. In the present review we discuss the evidence for NfL as a reliable biomarker for the early detection and management of MS. Moreover, we highlight the correlation between MRI and NfL, and ask whether they can be combined.
Collapse
Affiliation(s)
- Ebrahim Kouchaki
- MS Fellowship, Department of Neurology, School of Medicine, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Paramedical School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran
| |
Collapse
|