1
|
Yilmaz SN, Steiner K, Marksteiner J, Faserl K, Villunger M, Sarg B, Humpel C. From Organotypic Mouse Brain Slices to Human Alzheimer's Plasma Biomarkers: A Focus on Nerve Fiber Outgrowth. Biomolecules 2024; 14:1326. [PMID: 39456259 PMCID: PMC11506054 DOI: 10.3390/biom14101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory loss and progressive deterioration of cognitive functions. Being able to identify reliable biomarkers in easily available body fluids such as blood plasma is vital for the disease. To achieve this, we used a technique that applied human plasma to organotypic brain slice culture via microcontact printing. After a 2-week culture period, we performed immunolabeling for neurofilament and myelin oligodendrocyte glycoprotein (MOG) to visualize newly formed nerve fibers and oligodendrocytes. There was no significant change in the number of new nerve fibers in the AD plasma group compared to the healthy control group, while the length of the produced fibers significantly decreased. A significant increase in the number of MOG+ dots around these new fibers was detected in the patient group. According to our hypothesis, there are factors in the plasma of AD patients that affect the growth of new nerve fibers, which also affect the oligodendrocytes. Based on these findings, we selected the most promising plasma samples and conducted mass spectrometry using a differential approach and we identified three putative biomarkers: aldehyde-dehydrogenase 1A1, alpha-synuclein and protein S100-A4. Our method represents a novel and innovative approach for translating research findings from mouse models to human applications.
Collapse
Affiliation(s)
- Sakir Necat Yilmaz
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin 33130, Turkey
| | - Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Hall State Hospital, 6060 Hall in Tirol, Austria;
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Mathias Villunger
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, CCB-Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.F.); (M.V.); (B.S.)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.N.Y.); (K.S.)
| |
Collapse
|
2
|
Ponleitner M, Gatterer C, Bsteh G, Rath J, Altmann P, Berger T, Graf S, Sunder-Plassmann G, Rommer PS. Investigation of serum neurofilament light chain as a biomarker in Fabry disease. Sci Rep 2024; 14:23033. [PMID: 39362930 PMCID: PMC11449901 DOI: 10.1038/s41598-024-73537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Fabry disease (FD) constitutes a rare, X-linked lysosomal storage disorder affecting multiple organ systems, most notably heart, kidneys, and the central nervous system. Neurofilament light chains (NfL) have emerged as a prime candidate for a body fluid biomarker reflecting neuro-axonal injury. We aimed to evaluate its addition to the diagnostic and monitoring armamentarium in FD. Serum NfL concentrations (sNfL) were measured in 50 people with FD (PwFD) and 30 healthy control subjects (HC) using the Simoa© technology, followed by calculation of Z-scores adjusted for age and body mass index. In addition, clinical disease severity in PwFD was measured using the FOS-MSSI (Fabry outcome study - Mainz severity score index), which comprises clinical and paraclinical parameters. PwFD show elevated sNfL Z-scores compared to HC (PwFD: 1.12 [SD 1.5], HC: 0.01 [SD 1.2], p < 0.001). In PwFD, males showed higher sNfL Z-scores than females (1.75 [SD 1.5] vs. 0.73 [SD 1.4]). Importantly, sNfL Z-scores were increased in PwFD with ischemic white matter lesions of the CNS (1.5, SD 2.2 vs. 0.5, SD 2.9, p = 0.03). In our small cohort, sNfL Z-scores correlated fairly with FOS-MSSI (Kendall's-Tau [τ] = 0.25, p = 0.01), and, interestingly with serum creatinine (τ = 0.28, p = 0.005) and estimated glomerular filtration rate (eGFR, τ =-0.28, p = 0.005). Based on these exploratory results, sNfL might provide value as a biomarker of neuroaxonal damage in FD, possibly reflecting cerebrovascular injury. Additionally, the correlation of sNfL with renal function warrants further investigation.
Collapse
Affiliation(s)
- Markus Ponleitner
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria.
| | - Constantin Gatterer
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Jakob Rath
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Patrick Altmann
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Senta Graf
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Gere Sunder-Plassmann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Paulus Stefan Rommer
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Adelman JW, Sukowaty AT, Partridge KJ, Gawrys JE, Terhune SS, Ebert AD. Stabilizing microtubules aids neurite structure and disrupts syncytia formation in human cytomegalovirus-infected human forebrain neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608340. [PMID: 39229072 PMCID: PMC11370344 DOI: 10.1101/2024.08.16.608340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Human cytomegalovirus (HCMV) is a prolific human herpesvirus that infects most individuals by adulthood. While typically asymptomatic in adults, congenital infection can induce serious neurological symptoms including hearing loss, visual deficits, cognitive impairment, and microcephaly in 10-15% of cases. HCMV has been shown to infect most neural cells with our group recently demonstrating this capacity in stem cell-derived forebrain neurons. Infection of neurons induces deleterious effects on calcium dynamics and electrophysiological function paired with gross restructuring of neuronal morphology. Here, we utilize an iPSC-derived model of the human forebrain to demonstrate how HCMV infection induces syncytia, drives neurite retraction, and remodels microtubule networks to promote viral production and release. We establish that HCMV downregulates microtubule associated proteins at 14 days postinfection while simultaneously sparing other cytoskeletal elements, and this includes HCMV-driven alterations to microtubule stability. Further, we pharmacologically modulate microtubule dynamics using paclitaxel (stabilize) and colchicine (destabilize) to examine the effects on neurite structure, syncytial morphology, assembly compartment formation, and viral release. With paclitaxel, we found improvement of neurite outgrowth with a corresponding disruption to HCMV-induced syncytia formation and Golgi network disruptions but with limited impact on viral titers. Together, these data suggest that HCMV infection-induced disruption of microtubules in human cortical neurons can be partially mitigated with microtubule stabilization, suggesting a potential avenue for future neuroprotective therapeutic exploration.
Collapse
Affiliation(s)
- Jacob W Adelman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrew T Sukowaty
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kaitlyn J Partridge
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jessica E. Gawrys
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Marquette University and Medical College of Wisconsin Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
4
|
Mielke MM, Fowler NR. Alzheimer disease blood biomarkers: considerations for population-level use. Nat Rev Neurol 2024; 20:495-504. [PMID: 38862788 PMCID: PMC11347965 DOI: 10.1038/s41582-024-00989-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
In the past 5 years, we have witnessed the first approved Alzheimer disease (AD) disease-modifying therapy and the development of blood-based biomarkers (BBMs) to aid the diagnosis of AD. For many reasons, including accessibility, invasiveness and cost, BBMs are more acceptable and feasible for patients than a lumbar puncture (for cerebrospinal fluid collection) or neuroimaging. However, many questions remain regarding how best to utilize BBMs at the population level. In this Review, we outline the factors that warrant consideration for the widespread implementation and interpretation of AD BBMs. To set the scene, we review the current use of biomarkers, including BBMs, in AD. We go on to describe the characteristics of typical patients with cognitive impairment in primary care, who often differ from the patient populations used in AD BBM research studies. We also consider factors that might affect the interpretation of BBM tests, such as comorbidities, sex and race or ethnicity. We conclude by discussing broader issues such as ethics, patient and provider preference, incidental findings and dealing with indeterminate results and imperfect accuracy in implementing BBMs at the population level.
Collapse
Affiliation(s)
- Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Nicole R Fowler
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Center for Aging Research, Indianapolis, IN, USA
- Regenstrief Institute, Inc., Indianapolis, IN, USA
| |
Collapse
|
5
|
Behairi N, Samer A, Sahraoui L, Mataam DH, Trari R, Flissi B, Belguendouz H, Amir ZC, Touil-Boukoffa C. Neuroinflammation, neurodegeneration and alteration of spatial memory in BALB/c mice through ampicillin-induced gut dysbiosis; NOS2 and NFL involvement in a microbiota-gut-brain axis model. J Neuroimmunol 2024; 392:578374. [PMID: 38797060 DOI: 10.1016/j.jneuroim.2024.578374] [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: 02/19/2024] [Revised: 04/05/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
We aimed to investigate ampicillin (AMP) mechanisms in microbiota-gut-brain axis. We evaluated its effect on two gut and brain regions and behavioral performances. We administred AMP (1 g/l) to BALB/c mice for 21 days. Then, we analyzed body weigth change, stool consistency scoring, gut length, intestinal microbiota composition, nitric oxide synthase 2 (NOS2) expression and tissue integrity. We subsequently evaluated NOS2, GFAP, CD68 and NFL cerebral expression and spatial memory.Interestingly, our data showed gut microbiota disruption, NOS2 upregulation and tissue damage, associated to cerebral NOS2, GFAP, CD68 and NFL over-expression and behavioral alteration. Antiobiotic therapy should be prescribed with great caution.
Collapse
Affiliation(s)
- Nassima Behairi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Arezki Samer
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Lynda Sahraoui
- Laboratory of Animal Health and Production, Higher National Veterinary School of Issad-Abbes Oued-Smar, Algiers, Algeria
| | - Djehane Houria Mataam
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Ryad Trari
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Billel Flissi
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Houda Belguendouz
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria
| | - Zine-Charaf Amir
- Department of Anatomy and Pathological Cytology, University Hospital Center Mustapha Pacha, 1945 Pl. May 1st, Sidi M'Hamed, 16000 Algiers, Algeria
| | - Chafia Touil-Boukoffa
- University of Sciences and Technology Houari Boumediene (USTHB), Faculty of Biological Sciences, Cellular and Molecular Biology Laboratory, Cytokines and NO Synthases, Immunity and Pathogeny Team, El-Alia, BP 32, 16111 Algiers, Algeria.
| |
Collapse
|
6
|
Penny LK, Lofthouse R, Arastoo M, Porter A, Palliyil S, Harrington CR, Wischik CM. Considerations for biomarker strategies in clinical trials investigating tau-targeting therapeutics for Alzheimer's disease. Transl Neurodegener 2024; 13:25. [PMID: 38773569 PMCID: PMC11107038 DOI: 10.1186/s40035-024-00417-w] [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: 11/08/2023] [Accepted: 04/24/2024] [Indexed: 05/24/2024] Open
Abstract
The use of biomarker-led clinical trial designs has been transformative for investigating amyloid-targeting therapies for Alzheimer's disease (AD). The designs have ensured the correct selection of patients on these trials, supported target engagement and have been used to support claims of disease modification and clinical efficacy. Ultimately, this has recently led to approval of disease-modifying, amyloid-targeting therapies for AD; something that should be noted for clinical trials investigating tau-targeting therapies for AD. There is a clear overlap of the purpose of biomarker use at each stage of clinical development between amyloid-targeting and tau-targeting clinical trials. However, there are differences within the potential context of use and interpretation for some biomarkers in particular measurements of amyloid and utility of soluble, phosphorylated tau biomarkers. Given the complexities of tau in health and disease, it is paramount that therapies target disease-relevant tau and, in parallel, appropriate assays of target engagement are developed. Tau positron emission tomography, fluid biomarkers reflecting tau pathology and downstream measures of neurodegeneration will be important both for participant recruitment and for monitoring disease-modification in tau-targeting clinical trials. Bespoke design of biomarker strategies and interpretations for different modalities and tau-based targets should also be considered.
Collapse
Affiliation(s)
- Lewis K Penny
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Richard Lofthouse
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Mohammad Arastoo
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Andy Porter
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Soumya Palliyil
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- GT Diagnostics (UK) Ltd, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
- GT Diagnostics (UK) Ltd, Aberdeen, UK.
- TauRx Therapeutics Ltd, Aberdeen, UK.
| |
Collapse
|
7
|
Krause Neto W, Silva W, Oliveira T, Vilas Boas A, Ciena A, Caperuto ÉC, Gama EF. Ladder-based resistance training with the progression of training load altered the tibial nerve ultrastructure and muscle fiber area without altering the morphology of the postsynaptic compartment. Front Physiol 2024; 15:1371839. [PMID: 38694209 PMCID: PMC11061484 DOI: 10.3389/fphys.2024.1371839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Scientific evidence regarding the effect of different ladder-based resistance training (LRT) protocols on the morphology of the neuromuscular system is scarce. Therefore, the present study aimed to compare the morphological response induced by different LRT protocols in the ultrastructure of the tibial nerve and morphology of the motor endplate and muscle fibers of the soleus and plantaris muscles of young adult Wistar rats. Rats were divided into groups: sedentary control (control, n = 9), a predetermined number of climbs and progressive submaximal intensity (fixed, n = 9), high-intensity and high-volume pyramidal system with a predetermined number of climbs (Pyramid, n = 9) and lrt with a high-intensity pyramidal system to exhaustion (failure, n = 9). myelinated fibers and myelin sheath thickness were statistically larger in pyramid, fixed, and failure. myelinated axons were statistically larger in pyramid than in control. schwann cell nuclei were statistically larger in pyramid, fixed, and failure. microtubules and neurofilaments were greater in pyramid than in control. morphological analysis of the postsynaptic component of the plantar and soleus muscles did not indicate any significant difference. for plantaris, the type i myofibers were statistically larger in the pyramid and fixed compared to control. the pyramid, fixed, and failure groups for type ii myofibers had larger csa than control. for soleus, the type i myofibers were statistically larger in the pyramid than in control. pyramid and fixed had larger csa for type ii myofibers than control and failure. the pyramid and fixed groups showed greater mass progression delta than the failure. We concluded that the LRT protocols with greater volume and progression of accumulated mass elicit more significant changes in the ultrastructure of the tibial nerve and muscle hypertrophy without endplate changes.
Collapse
Affiliation(s)
- Walter Krause Neto
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Wellington Silva
- Depatment of Physical Education, Laboratory of Human Movement, Universidade São Judas Tadeu, São Paulo, Brazil
| | - Tony Oliveira
- Depatment of Physical Education, Laboratory of Human Movement, Universidade São Judas Tadeu, São Paulo, Brazil
| | - Alan Vilas Boas
- Depatment of Physical Education, Laboratory of Human Movement, Universidade São Judas Tadeu, São Paulo, Brazil
| | - Adriano Ciena
- Department of Physical Education, Laboratory of Morphology and Physical Activity, Universidade Estadual Paulista Júlio de Mesquita Filho, São Paulo, Brazil
| | - Érico Chagas Caperuto
- Depatment of Physical Education, Laboratory of Human Movement, Universidade São Judas Tadeu, São Paulo, Brazil
| | - Eliane Florencio Gama
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Yu F, Pituch KA, Maxfield M, Baena E, Geda YE, Pruzin JJ, Coon DW, Shaibi GQ. The associations between type 2 diabetes and plasma biomarkers of Alzheimer's disease in the Health and Aging Brain Study: Health Disparities (HABS-HD). PLoS One 2024; 19:e0295749. [PMID: 38558059 PMCID: PMC10984470 DOI: 10.1371/journal.pone.0295749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 11/28/2023] [Indexed: 04/04/2024] Open
Abstract
Alzheimer's disease (AD) affects Latinos disproportionately. One of the reasons underlying this disparity may be type 2 diabetes (T2D) that is a risk factor for AD. The purpose of this study was to examine the associations of T2D and AD blood biomarkers and the differences in these associations between Mexican Americans and non-Hispanic Whites. This study was a secondary analysis of baseline data from the observational Health and Aging Brain Study: Health Disparities (HABS-HD) that investigated factors underlying health disparities in AD in Mexican Americans in comparison to non-Hispanic Whites. HABS-HD participants were excluded if they had missing data or were large outliers (z-scores >|4|) on a given AD biomarker. Fasting blood glucose and glycosylated hemoglobin (HbA1c) levels were measured from clinical labs. T2D was diagnosed by licensed clinicians. Plasma amyloid-beta 42 and 40 (Aβ42/42) ratio, total tau (t-tau), and neurofilament light (NfL) were measured via ultra-sensitive Simoa assays. The sample sizes were 1,552 for Aβ42/40 ratio, 1,570 for t-tau, and 1,553 for NfL. Mexican Americans were younger (66.6±8.7 vs. 69.5±8.6) and had more female (64.9% female vs. 55.1%) and fewer years of schooling (9.5±4.6 vs. 15.6±2.5) than non-Hispanic Whites. Mexican Americans differed significantly from non-Hispanic Whites in blood glucose (113.5±36.6 vs. 99.2±17.0) and HbA1c (6.33±1.4 vs. 5.51±0.6) levels, T2D diagnosis (35.3% vs. 11.1%), as well as blood Aβ42/40 ratio (.051±.012 vs. .047±.011), t-tau (2.56±.95 vs. 2.33±.90), and NfL levels (16.3±9.5 vs. 20.3±10.3). Blood glucose, blood HbA1c, and T2D diagnosis were not related to Aβ42/40 ratio and t-tau but explained 3.7% of the variation in NfL (p < .001). Blood glucose and T2D diagnosis were not, while HbA1c was positively (b = 2.31, p < .001, β = 0.26), associated with NfL among Mexican Americans. In contrast, blood glucose, HbA1c, and T2D diagnosis were negatively (b = -0.09, p < .01, β = -0.26), not (b = 0.34, p = .71, β = 0.04), and positively (b = 3.32, p < .01, β = 0.33) associated with NfL, respectively in non-Hispanic Whites. To conclude, blood glucose and HbA1c levels and T2D diagnosis are associated with plasma NfL levels, but not plasma Aβ and t-tau levels. These associations differ in an ethnicity-specific manner and need to be further studied as a potential mechanism underlying AD disparities.
Collapse
Affiliation(s)
- Fang Yu
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Keenan A. Pituch
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Molly Maxfield
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Elsa Baena
- Clinical Neuropsychology Department, Barrow Neurological Institute, Phoenix, Arizona, United States of America
| | - Yonas E. Geda
- Department of Neurology and the Franke Neursciene Education Center, Barrow Neurological Institute, Phoenix, Arizona, United States of America
| | - Jeremy J. Pruzin
- Department of Neurology, Banner Alzheimer’s Institute, Phoenix, Arizona, United States of America
| | - David W. Coon
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | - Gabriel Q. Shaibi
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, United States of America
| | | |
Collapse
|
9
|
Gordon T. Brief Electrical Stimulation Promotes Recovery after Surgical Repair of Injured Peripheral Nerves. Int J Mol Sci 2024; 25:665. [PMID: 38203836 PMCID: PMC10779324 DOI: 10.3390/ijms25010665] [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: 10/13/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Injured peripheral nerves regenerate their axons in contrast to those in the central nervous system. Yet, functional recovery after surgical repair is often disappointing. The basis for poor recovery is progressive deterioration with time and distance of the growth capacity of the neurons that lose their contact with targets (chronic axotomy) and the growth support of the chronically denervated Schwann cells (SC) in the distal nerve stumps. Nonetheless, chronically denervated atrophic muscle retains the capacity for reinnervation. Declining electrical activity of motoneurons accompanies the progressive fall in axotomized neuronal and denervated SC expression of regeneration-associated-genes and declining regenerative success. Reduced motoneuronal activity is due to the withdrawal of synaptic contacts from the soma. Exogenous neurotrophic factors that promote nerve regeneration can replace the endogenous factors whose expression declines with time. But the profuse axonal outgrowth they provoke and the difficulties in their delivery hinder their efficacy. Brief (1 h) low-frequency (20 Hz) electrical stimulation (ES) proximal to the injury site promotes the expression of endogenous growth factors and, in turn, dramatically accelerates axon outgrowth and target reinnervation. The latter ES effect has been demonstrated in both rats and humans. A conditioning ES of intact nerve days prior to nerve injury increases axonal outgrowth and regeneration rate. Thereby, this form of ES is amenable for nerve transfer surgeries and end-to-side neurorrhaphies. However, additional surgery for applying the required electrodes may be a hurdle. ES is applicable in all surgeries with excellent outcomes.
Collapse
Affiliation(s)
- Tessa Gordon
- Division of Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON M4G 1X8, Canada
| |
Collapse
|
10
|
Kagerer SM, Awasthi S, Ripke S, Maceski A, Benkert P, Fall AB, Riederer P, Fischer P, Walitza S, Grünblatt E, Kuhle J, Unschuld PG. Polygenic risk for Alzheimer's disease is associated with neuroaxonal damage before onset of clinical symptoms. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12504. [PMID: 38213949 PMCID: PMC10776830 DOI: 10.1002/dad2.12504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION Establishing valid diagnostic strategies is a precondition for successful therapeutic intervention in Alzheimer's disease (AD). METHODS One hundred forty-four healthy 75-year-old participants from the Vienna-Transdanube-Aging longitudinal cohort study were tested for neuroaxonal damage by single molecular array (Simoa) plasma neurofilament light chain (NfL) levels at baseline, 30, 60, and 90 months, and onset of AD dementia. Individual risk for sporadic AD was estimated by continuous shrinkage polygenic risk score (PRS-CS, genome-wide association study). RESULTS Nineteen participants developed AD after a median of 60 months (interquartile range 30). In participants with AD, baseline NfL plasma levels correlated with PRS-CS (r = 0.75, p < 0.001; difference to controls: Fisher's r-to-z: z = 3.89, p < 0.001). PRS-CS combined with baseline plasma NfL predicted onset of AD (p < 0.01). DISCUSSION Our data suggest that polygenic risk for AD and plasma NfL closely interact years before onset of clinical symptoms. Peripheral NfL may serve as a diagnostic measure supporting early therapeutic intervention and secondary prevention in AD.
Collapse
Affiliation(s)
- Sonja M. Kagerer
- Department of Geriatric PsychiatryPsychiatric University Hospital Zurich (PUK)ZurichSwitzerland
- Institute for Regenerative Medicine (IREM)University of ZurichSchlierenSwitzerland
| | - Swapnil Awasthi
- Department of Psychiatry and PsychotherapyCharité‐UniversitätsmedizinBerlinGermany
| | - Stephan Ripke
- Department of Psychiatry and PsychotherapyCharité‐UniversitätsmedizinBerlinGermany
- Analytic and Translational Genetics UnitMassachusetts General HospitalBostonMassachusettsUSA
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Aleksandra Maceski
- Department of NeurologyUniversity Hospital and University of BaselBaselSwitzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB)Departments of Biomedicine and Clinical ResearchUniversity Hospital and University of BaselBaselSwitzerland
| | - Pascal Benkert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB)Departments of Biomedicine and Clinical ResearchUniversity Hospital and University of BaselBaselSwitzerland
- Departments of Clinical ResearchUniversity Hospital and University of BaselBaselSwitzerland
| | - Aïda B. Fall
- Geriatric Psychiatry ServiceUniversity Hospitals of Geneva (HUG)ThônexSwitzerland
- Department of PsychiatryUniversity of Geneva (UniGE)GenevaSwitzerland
| | - Peter Riederer
- Center of Mental HealthClinic and Policlinic of PsychiatryPsychosomatics and PsychotherapyUniversity Hospital of WürzburgWürzburgGermany
- Department of PsychiatryUniversity of Southern Denmark OdenseOdenseDenmark
| | - Peter Fischer
- Department of PsychiatrySocial Medicine Center East‐DonauspitalViennaAustria
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and PsychotherapyPsychiatric University Hospital ZurichUniversity of ZurichZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichSwiss Federal Institute of Technology and University of ZurichZurichSwitzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and PsychotherapyPsychiatric University Hospital ZurichUniversity of ZurichZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichSwiss Federal Institute of Technology and University of ZurichZurichSwitzerland
| | - Jens Kuhle
- Department of NeurologyUniversity Hospital and University of BaselBaselSwitzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB)Departments of Biomedicine and Clinical ResearchUniversity Hospital and University of BaselBaselSwitzerland
| | - Paul G. Unschuld
- Geriatric Psychiatry ServiceUniversity Hospitals of Geneva (HUG)ThônexSwitzerland
- Department of PsychiatryUniversity of Geneva (UniGE)GenevaSwitzerland
| |
Collapse
|
11
|
López-Erauskin J, Bravo-Hernandez M, Presa M, Baughn MW, Melamed Z, Beccari MS, Agra de Almeida Quadros AR, Arnold-Garcia O, Zuberi A, Ling K, Platoshyn O, Niño-Jara E, Ndayambaje IS, McAlonis-Downes M, Cabrera L, Artates JW, Ryan J, Hermann A, Ravits J, Bennett CF, Jafar-Nejad P, Rigo F, Marsala M, Lutz CM, Cleveland DW, Lagier-Tourenne C. Stathmin-2 loss leads to neurofilament-dependent axonal collapse driving motor and sensory denervation. Nat Neurosci 2024; 27:34-47. [PMID: 37996528 PMCID: PMC10842032 DOI: 10.1038/s41593-023-01496-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Jone López-Erauskin
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Mariana Bravo-Hernandez
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | | | - Michael W Baughn
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ze'ev Melamed
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Melinda S Beccari
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ana Rita Agra de Almeida Quadros
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Olatz Arnold-Garcia
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Neurosciences, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | | | - Karen Ling
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Oleksandr Platoshyn
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Elkin Niño-Jara
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - I Sandra Ndayambaje
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Melissa McAlonis-Downes
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Larissa Cabrera
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Jonathan W Artates
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Anita Hermann
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Martin Marsala
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Don W Cleveland
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
12
|
Barakovic M, Pizzolato M, Tax CMW, Rudrapatna U, Magon S, Dyrby TB, Granziera C, Thiran JP, Jones DK, Canales-Rodríguez EJ. Estimating axon radius using diffusion-relaxation MRI: calibrating a surface-based relaxation model with histology. Front Neurosci 2023; 17:1209521. [PMID: 37638307 PMCID: PMC10457121 DOI: 10.3389/fnins.2023.1209521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Axon radius is a potential biomarker for brain diseases and a crucial tissue microstructure parameter that determines the speed of action potentials. Diffusion MRI (dMRI) allows non-invasive estimation of axon radius, but accurately estimating the radius of axons in the human brain is challenging. Most axons in the brain have a radius below one micrometer, which falls below the sensitivity limit of dMRI signals even when using the most advanced human MRI scanners. Therefore, new MRI methods that are sensitive to small axon radii are needed. In this proof-of-concept investigation, we examine whether a surface-based axonal relaxation process could mediate a relationship between intra-axonal T2 and T1 times and inner axon radius, as measured using postmortem histology. A unique in vivo human diffusion-T1-T2 relaxation dataset was acquired on a 3T MRI scanner with ultra-strong diffusion gradients, using a strong diffusion-weighting (i.e., b = 6,000 s/mm2) and multiple inversion and echo times. A second reduced diffusion-T2 dataset was collected at various echo times to evaluate the model further. The intra-axonal relaxation times were estimated by fitting a diffusion-relaxation model to the orientation-averaged spherical mean signals. Our analysis revealed that the proposed surface-based relaxation model effectively explains the relationship between the estimated relaxation times and the histological axon radius measured in various corpus callosum regions. Using these histological values, we developed a novel calibration approach to predict axon radius in other areas of the corpus callosum. Notably, the predicted radii and those determined from histological measurements were in close agreement.
Collapse
Affiliation(s)
- Muhamed Barakovic
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, Wales, United Kingdom
- Signal Processing Laboratory 5 (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Marco Pizzolato
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Chantal M. W. Tax
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, Wales, United Kingdom
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, Netherlands
| | - Umesh Rudrapatna
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, Wales, United Kingdom
| | - Stefano Magon
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Tim B. Dyrby
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
- Danish Research Centre for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Cristina Granziera
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jean-Philippe Thiran
- Signal Processing Laboratory 5 (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Radiology Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- Centre d’Imagerie Biomédicale (CIBM), EPFL, Lausanne, Switzerland
| | - Derek K. Jones
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, Wales, United Kingdom
| | - Erick J. Canales-Rodríguez
- Signal Processing Laboratory 5 (LTS5), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
13
|
Ellen O, Ye S, Nheu D, Dass M, Pagnin M, Ozturk E, Theotokis P, Grigoriadis N, Petratos S. The Heterogeneous Multiple Sclerosis Lesion: How Can We Assess and Modify a Degenerating Lesion? Int J Mol Sci 2023; 24:11112. [PMID: 37446290 DOI: 10.3390/ijms241311112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous disease of the central nervous system that is governed by neural tissue loss and dystrophy during its progressive phase, with complex reactive pathological cellular changes. The immune-mediated mechanisms that promulgate the demyelinating lesions during relapses of acute episodes are not characteristic of chronic lesions during progressive MS. This has limited our capacity to target the disease effectively as it evolves within the central nervous system white and gray matter, thereby leaving neurologists without effective options to manage individuals as they transition to a secondary progressive phase. The current review highlights the molecular and cellular sequelae that have been identified as cooperating with and/or contributing to neurodegeneration that characterizes individuals with progressive forms of MS. We emphasize the need for appropriate monitoring via known and novel molecular and imaging biomarkers that can accurately detect and predict progression for the purposes of newly designed clinical trials that can demonstrate the efficacy of neuroprotection and potentially neurorepair. To achieve neurorepair, we focus on the modifications required in the reactive cellular and extracellular milieu in order to enable endogenous cell growth as well as transplanted cells that can integrate and/or renew the degenerative MS plaque.
Collapse
Affiliation(s)
- Olivia Ellen
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Danica Nheu
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| |
Collapse
|
14
|
Bircak-Kuchtova B, Chung HY, Wickel J, Ehler J, Geis C. Neurofilament light chains to assess sepsis-associated encephalopathy: Are we on the track toward clinical implementation? Crit Care 2023; 27:214. [PMID: 37259091 DOI: 10.1186/s13054-023-04497-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Sepsis is the most common cause of admission to intensive care units worldwide. Sepsis patients frequently suffer from sepsis-associated encephalopathy (SAE) reflecting acute brain dysfunction. SAE may result in increased mortality, extended length of hospital stay, and long-term cognitive dysfunction. The diagnosis of SAE is based on clinical assessments, but a valid biomarker to identify and confirm SAE and to assess SAE severity is missing. Several blood-based biomarkers indicating neuronal injury have been evaluated in sepsis and their potential role as early diagnosis and prognostic markers has been studied. Among those, the neuroaxonal injury marker neurofilament light chain (NfL) was identified to potentially serve as a prognostic biomarker for SAE and to predict long-term cognitive impairment. In this review, we summarize the current knowledge of biomarkers, especially NfL, in SAE and discuss a possible future clinical application considering existing limitations.
Collapse
Affiliation(s)
- Barbora Bircak-Kuchtova
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Ha-Yeun Chung
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany.
| | - Jonathan Wickel
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747, Jena, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| |
Collapse
|
15
|
Cheung SKK, Kwok J, Or PMY, Wong CW, Feng B, Choy KW, Chang RCC, Burbach JPH, Cheng ASL, Chan AM. Neuropathological signatures revealed by transcriptomic and proteomic analysis in Pten-deficient mouse models. Sci Rep 2023; 13:6763. [PMID: 37185447 PMCID: PMC10130134 DOI: 10.1038/s41598-023-33869-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
PTEN hamartoma tumour syndrome is characterised by mutations in the human PTEN gene. We performed transcriptomic and proteomic analyses of neural tissues and primary cultures from heterozygous and homozygous Pten-knockout mice. The somatosensory cortex of heterozygous Pten-knockout mice was enriched in immune response and oligodendrocyte development Gene Ontology (GO) terms. Parallel proteomic analysis revealed differentially expressed proteins (DEPs) related to dendritic spine development, keratinisation and hamartoma signatures. However, primary astrocytes (ASTs) from heterozygous Pten-knockout mice were enriched in the extracellular matrix GO term, while primary cortical neurons (PCNs) were enriched in immediate-early genes. In ASTs from homozygous Pten-knockout mice, cilium-related activity was enriched, while PCNs exhibited downregulation of forebrain neuron generation and differentiation, implying an altered excitatory/inhibitory balance. By integrating DEPs with pre-filtered differentially expressed genes, we identified the enrichment of traits of intelligence, cognitive function and schizophrenia, while DEPs in ASTs were significantly associated with intelligence and depression.
Collapse
Affiliation(s)
- Stanley K K Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Jacinda Kwok
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Penelope M Y Or
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Chi Wai Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Bo Feng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Kwong Wai Choy
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Raymond C C Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - J Peter H Burbach
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Andrew M Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China.
- Brain and Mind Institute, The Chinese University of Hong Kong, 4/F, Hui Yeung Shing Building, Hong Kong, SAR, China.
| |
Collapse
|
16
|
Ripamonti E, Edén A, Nilsson S, Sönnerborg A, Zetterberg H, Gisslén M. Longitudinal decline of plasma neurofilament light levels after antiretroviral initiation in people living with HIV. J Intern Med 2023; 293:445-456. [PMID: 36443917 DOI: 10.1111/joim.13594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND This retrospective follow-up study aims to investigate the dynamic longitudinal change of plasma neurofilament light (NfL) levels after antiretroviral therapy (ART) initiation in a cohort of people living with human immunodeficiency virus (HIV) (PWH). METHODS We tested a convenience sample of 116 patients from the NORTHIV study. Plasma NfL levels-measured using Single molecule array (Simoa) technology-as well as other laboratory parameters were collected at baseline, weeks 4, 48, 96, and 144. Linear mixed-effects models were estimated to evaluate longitudinal change over time. Baseline CD4+ T-cell levels, CDC classification, and HIV RNA levels were considered. Models were adjusted by age, sex, treatment regimen, and baseline serum creatinine levels. RESULTS Plasma NfL levels were higher at baseline and also declined faster during the follow-up for participants with CD4+ count <100 cells/µl compared with >100 cells/µl. No significant difference was found between the CD4+ strata 100-199 and 200-499/µl. Participants with CDC classification stages B and C had higher levels of plasma NfL at baseline, as well as faster decline compared with participants with stage A. No significant main effects or change over time was found in baseline HIV RNA levels, treatment regimen, or sex. CONCLUSION Plasma NfL is a sensitive biomarker to assess ongoing central nervous system injury in PWH. Plasma NfL concentrations decline relatively fast following ART initiation and then stabilize after 48 weeks. Plasma NfL concentrations are associated with CD4+ count and stage of HIV disease. No correlations were seen with different ART regimens.
Collapse
Affiliation(s)
- Enrico Ripamonti
- Milan Center for Neuroscience, University of Milan-Bicocca, Milan, Italy.,Department of Economics and Management, University of Brescia, Brescia, Italy
| | - Arvid Edén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Sönnerborg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine ANA Futura Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
17
|
Kaur S, Zhang X, Patel S, Rodriguez YA, Luther KJ, Alghafli G, Lang RM, Abrams CK, Dobrowsky RT. Pharmacologic Targeting of the C-Terminus of Heat Shock Protein 90 Improves Neuromuscular Function in Animal Models of Charcot Marie Tooth X1 Disease. ACS Pharmacol Transl Sci 2023; 6:306-319. [PMID: 36798471 PMCID: PMC9926526 DOI: 10.1021/acsptsci.2c00223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Indexed: 01/22/2023]
Abstract
Charcot-Marie-Tooth X1 (CMTX1) disease is an inherited peripheral neuropathy that arises from loss-of-function mutations in the protein connexin 32 (Cx32). CMTX1 currently lacks a pharmacologic approach toward disease management, and we have previously shown that modulating the expression of molecular chaperones using novologue therapy may provide a viable disease-modifying approach to treat metabolic and demyelinating neuropathies. Cemdomespib is an orally bioavailable novologue that manifests neuroprotective activity by modulating the expression of heat shock protein 70 (Hsp70). We examined if 1 to 5 months of daily cemdomespib therapy may improve neuropathic symptoms in three mouse models of CMTX1 (Cx32 deficient (Cx32def), T55I-Cx32def, and R75W-Cx32 mice). Daily drug therapy significantly improved motor nerve conduction velocity (MNCV) and grip strength in all three models, but the compound muscle action potential was only improved in Cx32def mice. Drug efficacy required Hsp70 as improvements in MNCV, and the grip strength was abrogated in Cx32def × Hsp70 knockout mice. Five months of novologue therapy was associated with improved neuromuscular junction morphology, femoral motor nerve myelination, reduction in foamy macrophages, and a decrease in Schwann cell c-jun levels. To determine if c-jun may be downstream of Hsp70 and necessary for drug efficacy, c-jun expression was specifically deleted in Schwann cells of Cx32def mice. While the deletion of c-jun worsened the neuropathy, cemdomespib therapy remained effective in improving MNCV and grip strength. Our data show that cemdomespib therapy improves CMTX1-linked neuropathy in an Hsp70-dependent but a c-jun-independent manner and without regard to the nature of the underlying Cx32 mutation.
Collapse
Affiliation(s)
- Sukhmanjit Kaur
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Xinyue Zhang
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Sugandha Patel
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Yssa A. Rodriguez
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Kylie J. Luther
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Ghufran Alghafli
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Ryan M. Lang
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| | - Charles K. Abrams
- Department
of Neurology and Rehabilitation and Biomedical Engineering, College
of Medicine, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Rick T. Dobrowsky
- Department
of Pharmacology and Toxicology, University
of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
18
|
Rådestig MA, Skoog J, Zetterberg H, Skillbäck T, Zettergren A, Sterner TR, Fässberg MM, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Subtle Differences in Cognition in 70-Year-Olds with Elevated Cerebrospinal Fluid Neurofilament Light and Neurogranin: A H70 Cross-Sectional Study. J Alzheimers Dis 2023; 91:291-303. [PMID: 36617786 PMCID: PMC9881027 DOI: 10.3233/jad-220452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Most research on cerebrospinal fluid (CSF) neurofilament light protein (NfL) as a marker for neurodegeneration and neurogranin (Ng) for synaptic dysfunction has largely focused on clinical cohorts rather than population-based samples. OBJECTIVE We hypothesized that increased CSF levels of NfL and Ng are associated with subtle cognitive deficits in cognitively unimpaired (CU) older adults. METHODS The sample was derived from the Gothenburg H70 Birth Cohort Studies and comprised 258 CU 70-year-olds, with a Clinical Dementia Rating score of zero. All participants underwent extensive cognitive testing. CSF levels of NfL and Ng, as well as amyloid β1 - 42, total tau, and phosphorylated tau, were measured. RESULTS Participants with high CSF NfL performed worse in one memory-based test (Immediate recall, p = 0.013) and a language test (FAS, p = 0.016). Individuals with high CSF Ng performed worse on the memory-based test Supra Span (p = 0.035). When stratified according to CSF tau and Aβ42 concentrations, participants with high NfL and increased tau performed worse on a memory test than participants normal tau concentrations (Delayed recall, p = 0.003). In participants with high NfL, those with pathologic Aβ42 concentrations performed worse on the Delayed recall memory (p = 0.044). In the high Ng group, participants with pathological Aβ42 concentrations had lower MMSE scores (p = 0.027). However, in regression analysis we found no linear correlations between CSF NfL or CSF Ng in relation to cognitive tests when controlled for important co-variates. CONCLUSION Markers of neurodegeneration and synaptic pathology might be associated with subtle signs of cognitive decline in a population-based sample of 70-year-olds.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UCL Institute of Neurology, Queen Square, London, UK
- The UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tobias Skillbäck
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Therese Rydberg Sterner
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Madeleine Mellqvist Fässberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Memory Disorders Clinic, Theme Inflammation and Aging, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Clinical Geriatric, Karolinska Institute, Stockholm, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychosis Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
19
|
Yuan A, Nixon RA. Posttranscriptional regulation of neurofilament proteins and tau in health and disease. Brain Res Bull 2023; 192:115-127. [PMID: 36441047 PMCID: PMC9907725 DOI: 10.1016/j.brainresbull.2022.10.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 01/16/2023]
Abstract
Neurofilament and tau proteins are neuron-specific cytoskeletal proteins that are enriched in axons, regulated by many of the same protein kinases, interact physically, and are the principal constituents of neurofibrillary lesions in major adult-onset dementias. Both proteins share functions related to the modulation of stability and functions of the microtubule network in axons, axonal transport and scaffolding of organelles, long-term synaptic potentiation, and learning and memory. Expression of these proteins is regulated not only at the transcriptional level but also through posttranscriptional control of pre-mRNA splicing, mRNA stability, transport, localization, local translation and degradation. Current evidence suggests that posttranscriptional determinants of their levels are usually regulated by RNA-binding proteins and microRNAs primarily through 3'-untranslated regions of neurofilament and tau mRNAs. Dysregulations of neurofilament and tau expression caused by mutations or pathologies of RNA-binding proteins such as TDP43, FUS and microRNAs are increasingly recognized in association with varied neurological disorders. In this review, we summarize the current understanding of posttranscriptional control of neurofilament and tau by examining the posttranscriptional regulation of neurofilament and tau by RNA-binding proteins and microRNAs implicated in health and diseases.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA; Department of Psychiatry, New York University Langone Health, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA,Department of Psychiatry, New York University Langone Health, New York, NY 10016, USA,Department of Cell Biology, New York University Langone Health, New York, NY 10016, USA,NYU Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA,Correspondence to: Center for Dementia Research, Nathan Kline Institute, New York University Langone Health, New York, NY 10016, USA, (A. Yuan), (R.A. Nixon)
| |
Collapse
|
20
|
Cerebrospinal Fluid Biomarkers in iNPH: A Narrative Review. Diagnostics (Basel) 2022; 12:diagnostics12122976. [PMID: 36552981 PMCID: PMC9777226 DOI: 10.3390/diagnostics12122976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is a neurological syndrome characterized by the clinical triad of gait disorder, cognitive impairment and urinary incontinence. It has attracted interest because of the possible reversibility of symptoms, especially with timely treatment. The main pathophysiological theory is based on a vicious circle of disruption in circulation of cerebrospinal fluid (CSF) that leads to the deceleration of its absorption. Data regarding CSF biomarkers in iNPH are contradictory and no definite CSF biomarker profile has been recognized as in Alzheimer's disease (AD), which often co-exists with iNPH. In this narrative review, we investigated the literature regarding CSF biomarkers in iNPH, both the established biomarkers total tau protein (t-tau), phosphorylated tau protein (p-tau) and amyloid peptide with 42 amino acids (Aβ42), and other molecules, which are being investigated as emerging biomarkers. The majority of studies demonstrate differences in CSF concentrations of Aβ42 and tau-proteins (t-tau and p-tau) among iNPH patients, healthy individuals and patients with AD and vascular dementia. iNPH patients present with lower CSF Aβ42 and p-tau concentrations than healthy individuals and lower t-tau and p-tau concentrations than AD patients. This could prove helpful for improving diagnosis, differential diagnosis and possibly prognosis of iNPH patients.
Collapse
|
21
|
Lynch M, Pham W, Sinclair B, O’Brien TJ, Law M, Vivash L. Perivascular spaces as a potential biomarker of Alzheimer's disease. Front Neurosci 2022; 16:1021131. [PMID: 36330347 PMCID: PMC9623161 DOI: 10.3389/fnins.2022.1021131] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/23/2022] [Indexed: 07/20/2023] Open
Abstract
Alzheimer's disease (AD) is a highly damaging disease that affects one's cognition and memory and presents an increasing societal and economic burden globally. Considerable research has gone into understanding AD; however, there is still a lack of effective biomarkers that aid in early diagnosis and intervention. The recent discovery of the glymphatic system and associated Perivascular Spaces (PVS) has led to the theory that enlarged PVS (ePVS) may be an indicator of AD progression and act as an early diagnostic marker. Visible on Magnetic Resonance Imaging (MRI), PVS appear to enlarge when known biomarkers of AD, amyloid-β and tau, accumulate. The central goal of ePVS and AD research is to determine when ePVS occurs in AD progression and if ePVS are causal or epiphenomena. Furthermore, if ePVS are indeed causative, interventions promoting glymphatic clearance are an attractive target for research. However, it is necessary first to ascertain where on the pathological progression of AD ePVS occurs. This review aims to examine the knowledge gap that exists in understanding the contribution of ePVS to AD. It is essential to understand whether ePVS in the brain correlate with increased regional tau distribution and global or regional Amyloid-β distribution and to determine if these spaces increase proportionally over time as individuals experience neurodegeneration. This review demonstrates that ePVS are associated with reduced glymphatic clearance and that this reduced clearance is associated with an increase in amyloid-β. However, it is not yet understood if ePVS are the outcome or driver of protein accumulation. Further, it is not yet clear if ePVS volume and number change longitudinally. Ultimately, it is vital to determine early diagnostic criteria and early interventions for AD to ease the burden it presents to the world; ePVS may be able to fulfill this role and therefore merit further research.
Collapse
Affiliation(s)
- Miranda Lynch
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - William Pham
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Benjamin Sinclair
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Radiology, Alfred Health, Melbourne, VIC, Australia
- Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, VIC, Australia
| | - Lucy Vivash
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
22
|
Hood CJ, Hendren NS, Pedretti R, Roth LR, Saelices L, Grodin JL. Update on Disease-Specific Biomarkers in Transthyretin Cardiac Amyloidosis. Curr Heart Fail Rep 2022; 19:356-363. [PMID: 35930129 PMCID: PMC10132942 DOI: 10.1007/s11897-022-00570-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
PURPOSE OF REVIEW Transthyretin cardiac amyloidosis (ATTR-CM) is an infiltrative cardiomyopathy and an increasingly recognized cause of morbidity and mortality. There remains substantial delay between initial symptoms and diagnosis. With the recent emergence of various targeted therapies proven to reduce morbidity and mortality, there is an imperative to diagnose subclinical disease. Biomarkers may be well-suited for this role. RECENT FINDINGS Conventional markers of heart failure, such as natriuretic peptides and cardiac troponins, and estimated glomerular filtration rate are associated with risk in ATTR-CM. Circulating transthyretin (TTR) levels parallel TTR kinetic stability, correlate with disease severity, and may serve as indirect markers of ATTR-CM disease activity and response to targeted treatment. There is also growing evidence for the correlation of TTR to retinol-binding protein 4, a biomarker which independently associates with this disease. The rate-limiting step for ATTR pathogenesis is dissociation of the TTR homotetramer, which may be quantified using subunit exchange to allow for early risk assessment, prognostication, and assessment of treatment response. The protein species that result from the dissociation and misfolding of TTR are known as nonnative transthyretin (NNTTR). NNTTR is quantifiable via peptide probes and is a specific biomarker whose reduction is positively correlated with improvement in neuropathic ATTR amyloidosis. Neurofilament light chain (NfL) is released into the blood after axonal damage and correlates with neuropathic ATTR amyloidosis, but its clinical use in ATTR-CM is uncertain. Conventional markers of heart failure, transthyretin, retinol-binding protein 4, transthyretin kinetic stability, nonnative transthyretin, peptide probes, and neurofilament light chain have potential as biomarkers to enable early, subclinical diagnosis in patients with transthyretin cardiac amyloidosis.
Collapse
Affiliation(s)
- Caleb J Hood
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Parkland Health and Hospital System, Dallas, TX, USA
| | - Nicholas S Hendren
- Parkland Health and Hospital System, Dallas, TX, USA
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Ste. E5.310F, Dallas, TX, 75390-8830, USA
| | - Rose Pedretti
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lori R Roth
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Ste. E5.310F, Dallas, TX, 75390-8830, USA
| | - Lorena Saelices
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin L Grodin
- Parkland Health and Hospital System, Dallas, TX, USA.
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Ste. E5.310F, Dallas, TX, 75390-8830, USA.
| |
Collapse
|
23
|
Neuro-Axonal Damage and Alteration of Blood–Brain Barrier Integrity in COVID-19 Patients. Cells 2022; 11:cells11162480. [PMID: 36010557 PMCID: PMC9406414 DOI: 10.3390/cells11162480] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 01/08/2023] Open
Abstract
Neurofilament light chain (NfL) is a specific biomarker of neuro-axonal damage. Matrix metalloproteinases (MMPs) are zinc-dependent enzymes involved in blood–brain barrier (BBB) integrity. We explored neuro-axonal damage, alteration of BBB integrity and SARS-CoV-2 RNA presence in COVID-19 patients with severe neurological symptoms (neuro-COVID) as well as neuro-axonal damage in COVID-19 patients without severe neurological symptoms according to disease severity and after recovery, comparing the obtained findings with healthy donors (HD). Overall, COVID-19 patients (n = 55) showed higher plasma NfL levels compared to HD (n = 31) (p < 0.0001), especially those who developed ARDS (n = 28) (p = 0.0005). After recovery, plasma NfL levels were still higher in ARDS patients compared to HD (p = 0.0037). In neuro-COVID patients (n = 12), higher CSF and plasma NfL, and CSF MMP-2 levels in ARDS than non-ARDS group were observed (p = 0.0357, p = 0.0346 and p = 0.0303, respectively). SARS-CoV-2 RNA was detected in four CSF and two plasma samples. SARS-CoV-2 RNA detection was not associated to increased CSF NfL and MMP levels. During COVID-19, ARDS could be associated to CNS damage and alteration of BBB integrity in the absence of SARS-CoV-2 RNA detection in CSF or blood. CNS damage was still detectable after discharge in blood of COVID-19 patients who developed ARDS during hospitalization.
Collapse
|
24
|
Carvalho DZ, St. Louis EK, Przybelski SA, Morgenthaler TI, Machulda MM, Boeve BF, Petersen RC, Jack CR, Graff-Radford J, Vemuri P, Mielke MM. Sleepiness in Cognitively Unimpaired Older Adults Is Associated With CSF Biomarkers of Inflammation and Axonal Integrity. Front Aging Neurosci 2022; 14:930315. [PMID: 35898322 PMCID: PMC9309557 DOI: 10.3389/fnagi.2022.930315] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Sleepiness has been associated with cognitive decline and dementia in the elderly. Older adults with excessive daytime sleepiness appear to be more vulnerable to longitudinal amyloid PET accumulation before the onset of the dementia. However, it remains unclear whether sleepiness is similarly associated with other biomarkers of Alzheimer's disease (AD), axonal integrity, and inflammation, which may also contribute to neurodegeneration and cognitive decline. Methods In this cross-sectional analysis, we identified 260 cognitively unimpaired adults (>60 years) from the Mayo Clinic Study of Aging, a population-based cohort from Olmsted County (MN), who underwent CSF quantification of AD biomarkers (Aβ42, p-tau, p-tau/Aβ42) in addition to at least one of the following biomarkers [neurofilament light chain (NfL) interleukin-6 (IL-6), IL-10, and tumor necrosis factor-α (TNF-α)]. We fit linear regression models to assess associations between sleepiness, as measured by the Epworth Sleepiness Scale (ESS), and CSF biomarkers, controlling for age, sex, APOε4 status, body mass index, hypertension, dyslipidemia, and prior diagnosis of obstructive sleep apnea. Results Higher ESS scores were associated with higher CSF IL-6 and NfL, but not with the other CSF biomarkers. For every ESS score point increase, there was a 0.009 ([95% CI 0.001-0.016], p = 0.033) increase in the log of IL-6 and 0.01 ([95% CI 0.002-0.018], p = 0.016) increase in the log of NfL. A sensitivity analysis showed an association between ESS scores and log of p-tau/Aβ42 only in participants with an abnormal ratio (>0.023), highly predictive of amyloid positivity. For every ESS score point increase, there was a 0.006 ([95% CI 0.001-0.012], p = 0.021) increase in the log of CSF p-tau/Aβ42. Conclusion Sleepiness was associated with greater CSF IL-6 and NfL levels, which could contribute to neurodegeneration or alternatively cause sleepiness. Higher NfL levels may result from sleep disruption and/or contribute to sleepiness via disturbed connectivity or damage to wake-promoting centers. Associations between sleepiness and p-tau/Aβ42 in participants with abnormal ratio suggest that amyloid positivity contributes to vulnerability to sleep disturbance, which may further amyloid accumulation in a feed-forward loop process. Prospective studies of these markers are needed to determine cause-effect relationships between these associations.
Collapse
Affiliation(s)
- Diego Z. Carvalho
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Sleep Medicine, Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Erik K. St. Louis
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Sleep Medicine, Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Scott A. Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Timothy I. Morgenthaler
- Center for Sleep Medicine, Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Mary M. Machulda
- Department of Psychology, Mayo Clinic, Rochester, MN, United States
| | - Bradley F. Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Sleep Medicine, Division of Pulmonary and Critical Care, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | | | | | - Michelle M. Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
25
|
Beydoun MA, Noren Hooten N, Weiss J, Beydoun HA, Hossain S, Evans MK, Zonderman AB. Plasma neurofilament light and its association with all-cause mortality risk among urban middle-aged men and women. BMC Med 2022; 20:218. [PMID: 35692046 PMCID: PMC9190073 DOI: 10.1186/s12916-022-02425-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/31/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Neurofilament light chain (NfL) is released into the blood during neuronal damage. NfL is linked to mortality in neurological disorders, remaining unexplored in population studies. We investigated whether initial (v1) and annualized change (δ) in plasma NfL can predict all-cause mortality in middle-aged dementia-free urban adults. METHODS Longitudinal data were from 694 participants in the Healthy Aging in Neighborhoods of Diversity Across the Life Span study (HANDLS, mean agev1: 47.8 years, 42% male, 55.8% African American). Plasma NfL was measured prospectively at three visits. Analyses included Cox proportional hazards models for all-cause mortality risk and 4-way decomposition testing for interaction and mediation. RESULTS Unlike men, women exhibited a direct association between δNfL (above vs. below median) and all-cause mortality risk in both the minimally (HR = 3.91, 95% CI 1.10-13.9, p = 0.036) and fully adjusted models (HR = 4.92, 95% CI 1.26-19.2, p = 0.022), and for δNfL (per unit increase) in the full model (HR = 1.65, 95% CI 1.04-2.61, p = 0.034). In both models, and among women, 1 standard deviation of NfLv1 was associated with an increased all-cause mortality risk (reduced model: HR = 2.01, 95% CI 1.24-3.25, p = 0.005; full model: HR = 1.75, 95% CI 1.02-2.98, p = 0.041). Only few interactions were detected for cardio-metabolic risk factors. Notably, NfLv1 was shown to be a better prognostic indicator at normal hsCRP values among women, while HbA1c and δNfL interacted synergistically to determine mortality risk, overall. CONCLUSIONS These findings indicate that plasma NfL levels at baseline and over time can predict all-cause mortality in women and interacts with hsCRP and HbA1c to predict that risk.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA.
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Jordan Weiss
- Department of Demography, University of California, Berkeley, Berkeley, CA, USA
| | - Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA
| | - Sharmin Hossain
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, 251 Bayview Blvd., Suite 100, Room #: 04B118, Baltimore, MD, 21224, USA
| |
Collapse
|
26
|
Kahouadji S, Bouillon-Minois JB, Oris C, Durif J, Pereira B, Pinguet J, Rozand A, Schmidt J, Sapin V, Bouvier D. Evaluation of serum neurofilament light in the early management of mTBI patients. Clin Chem Lab Med 2022; 60:1234-1241. [PMID: 35511901 DOI: 10.1515/cclm-2022-0173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/20/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Serum S100B allows a one-third reduction of computed tomography (CT) scans performed for mild traumatic brain injury (mTBI) patients. In this study, we evaluated the diagnostic performance of serum NF-L in the detection of intracranial lesions induced by mTBI. METHODS One hundred seventy-nine adult mTBI patients presenting to the emergency department of Clermont-Ferrand University Hospital with a Glasgow Coma Scale (GCS) score of 14-15 were included. S100B assays were performed for clinical routine while NF-L samples were stored at -80 °C until analysis. CT scans were performed for patients with S100B levels above the decision threshold of 0.10 μg/L. Later, NF-L and S100B levels were compared to CT scan findings to evaluate the biomarkers' performances. RESULTS The area under the ROC curve (AUC) evaluating the diagnostic ability in the prediction of intracranial lesions was 0.72 (95% CI; 0.58-0.87) for S100B and 0.58 (95% CI; 0.45-0.71) for NF-L, the specificities (at a threshold allowing a 100% sensitivity) were 35.7% for S100B, and 28% for NF-L (p=0.096). AUCs of NF-L and S100B for the identification of patients with neurological disorders were statistically different (p<0.001). The AUCs were 0.87 (95% CI; 0.82-0.93) for NF-L and 0.57 (95% CI; 0.48-0.66) for S100B. There was a poor correlation between NF-L and S100B, and NF-L levels were correlated to patients' age (Spearman coefficient of 0.79). CONCLUSIONS NF-L showed poor performances in the early management of mTBI patients. NF-L levels are strongly correlated to neurodegeneration, whether physiological, age-related, or pathological.
Collapse
Affiliation(s)
- Samy Kahouadji
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | | | - Charlotte Oris
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France.,Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Julie Durif
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Jérémy Pinguet
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Agathe Rozand
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Jeannot Schmidt
- Adult Emergency Department, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Sapin
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France.,Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Damien Bouvier
- Biochemistry and Molecular Genetic Department, CHU Clermont-Ferrand, Clermont-Ferrand, France.,Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| |
Collapse
|
27
|
Bendstrup N, Hejl AM, Salvesen L. Neurofilament Light Chain Levels in Frontotemporal Dementia and Progressive Supranuclear Palsy: A Systematic Review. J Alzheimers Dis 2022; 87:131-140. [PMID: 35275542 DOI: 10.3233/jad-215616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND It can be challenging to discriminate between progressive supranuclear palsy (PSP) and frontotemporal dementia (FTD). However, a correct diagnosis is a precondition for targeted treatment strategies and proper patient counseling. There has been a growing interest to identify cerebrospinal fluid (CSF) biomarkers, including neurofilament light chain (NfL). OBJECTIVE This systematic review evaluates the existing literature on neurofilament light in CSF aiming to validate its utility for differentiating FTD from PSP. METHODS A systematic literature search was conducted. A broad range of synonyms for PSP, NfL, and FTD as well as associated MeSH terms, were combined and used as keywords when searching. Relevant data were extracted and assessed for risk of bias. RESULTS Nine studies including a total of 671 patients with FTD, 254 patients with PSP, 523 healthy controls, and 1,771 patients with other disorders were included in the review. Four studies found a significantly higher level of CSF NfL in FTD (n = 445) compared to PSP (n = 124); however, in three of these studies the difference was only significant in certain FTD variants. Four studies found no significant difference in CSF NfL between PSP (n = 98) and FTD (n = 248). One study found a significantly higher level of NfL in PSP (n = 33) compared to FTD (n = 16). CONCLUSION In the majority of patients in the studies included in this review, a higher level of NfL in CSF was found in patients with FTD compared to patients with PSP; however, results were inconsistent and prospective studies including large study cohorts are needed.
Collapse
Affiliation(s)
- Nathalie Bendstrup
- Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anne-Mette Hejl
- Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Lisette Salvesen
- Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
28
|
Krause Neto W, Gama EF, Silva WDA, de Oliveira TVA, Vilas Boas AEDS, Ciena AP, Anaruma CA, Caperuto ÉC. The sciatic and radial nerves seem to adapt similarly to different ladder-based resistance training protocols. Exp Brain Res 2022; 240:887-896. [DOI: 10.1007/s00221-021-06295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/16/2021] [Indexed: 11/25/2022]
|
29
|
Leuzy A, Mattsson‐Carlgren N, Palmqvist S, Janelidze S, Dage JL, Hansson O. Blood-based biomarkers for Alzheimer's disease. EMBO Mol Med 2022; 14:e14408. [PMID: 34859598 PMCID: PMC8749476 DOI: 10.15252/emmm.202114408] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/01/2022] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent a mounting public health challenge. As these diseases are difficult to diagnose clinically, biomarkers of underlying pathophysiology are playing an ever-increasing role in research, clinical trials, and in the clinical work-up of patients. Though cerebrospinal fluid (CSF) and positron emission tomography (PET)-based measures are available, their use is not widespread due to limitations, including high costs and perceived invasiveness. As a result of rapid advances in the development of ultra-sensitive assays, the levels of pathological brain- and AD-related proteins can now be measured in blood, with recent work showing promising results. Plasma P-tau appears to be the best candidate marker during symptomatic AD (i.e., prodromal AD and AD dementia) and preclinical AD when combined with Aβ42/Aβ40. Though not AD-specific, blood NfL appears promising for the detection of neurodegeneration and could potentially be used to detect the effects of disease-modifying therapies. This review provides an overview of the progress achieved thus far using AD blood-based biomarkers, highlighting key areas of application and unmet challenges.
Collapse
Affiliation(s)
- Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Niklas Mattsson‐Carlgren
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Department of NeurologySkåne University HospitalLundSweden
- Wallenberg Centre for Molecular MedicineLund UniversityLundSweden
| | - Sebastian Palmqvist
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Shorena Janelidze
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | - Jeffrey L Dage
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisINUSA
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalLundSweden
| |
Collapse
|
30
|
Translocator Protein Ligand PIGA1138 Reduces Disease Symptoms and Severity in Experimental Autoimmune Encephalomyelitis Model of Primary Progressive Multiple Sclerosis. Mol Neurobiol 2022; 59:1744-1765. [PMID: 35018577 DOI: 10.1007/s12035-022-02737-2] [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: 10/30/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system (CNS) caused by CNS infiltration of peripheral immune cells, immune-mediated attack of the myelin sheath, neuroinflammation, and/or axonal/neuronal dysfunctions. Some drugs are available to cope with relapsing-remitting MS (RRMS) but there is no therapy for the primary progressive MS (PPMS). Because growing evidence supports a regulatory role of the translocator protein (TSPO) in neuroinflammatory, demyelinating, and neurodegenerative processes, we investigated the therapeutic potential of phenylindolyilglyoxylamydes (PIGAs) TSPO ligands in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) mice mimicking the human PPMS. MOG-EAE C57Bl/6-mice were treated by TSPO ligands PIGA839, PIGA1138, or the vehicle. Several methods were combined to evaluate PIGAs-TSPO ligand effects on MOG-EAE symptoms, CNS infiltration by immune cells, demyelination, and axonal damages. PIGA1138 (15 mg/kg) drastically reduced MOG-EAE mice clinical scores, ameliorated motor dysfunctions assessed with the Catwalk device, and counteracted MOG-EAE-induced demyelination by preserving Myelin basic protein (MBP) expression in the CNS. Furthermore, PIGA1138-treatment prevented EAE-evoked decreased neurofilament-200 expression in spinal and cerebellar axons. Moreover, PIGA1138 inhibited peripheral immune-CD45 + cell infiltration in the CNS, suggesting that it may control inflammatory mechanisms involved in PPMS. Concordantly, PIGA1138 enhanced anti-inflammatory interleukin-10 serum level in MOG-EAE mice. PIGA1138-treatment, which increased neurosteroid allopregnanolone production, ameliorated all pathological biomarkers, while PIGA839, unable to activate neurosteroidogenesis in vivo, exerted only moderate/partial effects in MOG-EAE mice. Altogether, our results suggest that PIGA1138-based treatment may represent an interesting possibility to be explored for the innovation of effective therapies against PPMS.
Collapse
|
31
|
Zhao Y, Arceneaux L, Culicchia F, Lukiw WJ. Neurofilament Light (NF-L) Chain Protein from a Highly Polymerized Structural Component of the Neuronal Cytoskeleton to a Neurodegenerative Disease Biomarker in the Periphery. HSOA JOURNAL OF ALZHEIMER'S & NEURODEGENERATIVE DISEASES 2021; 7:056. [PMID: 34881359 PMCID: PMC8651065 DOI: 10.24966/and-9608/100056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurofilaments (NFs) are critical scaffolding components of the axoskeleton of healthy neurons interacting directly with multiple synaptic-phosphoproteins to support and coordinate neuronal cell shape, cytoarchitecture, synaptogenesis and neurotransmission. While neuronal presynaptic proteins such as synapsin-2 (SYN II) degrade rapidly via the ubiquitin-proteasome pathway, a considerably more stable neurofilament light (NF-L) chain protein turns over much more slowly, and in several neurological diseases is accompanied by a pathological shift from an intracellular neuronal cytoplasmic location into various biofluid compartments. NF-L has been found to be significantly elevated in peripheral biofluids in multiple neurodegenerative disorders, however it is not as widely appreciated that NF-L expression within neurons undergoing inflammatory neurodegeneration exhibit a significant down-regulation in these neuron-specific intermediate-filament components. Down-regulated NF-L in neurons correlates well with the observed axonal and neuronal atrophy, neurite deterioration and synaptic disorganization in tissues affected by Alzheimer's disease (AD) and other progressive, age-related neurological diseases. This Review paper: (i) will briefly assess the remarkably high number of neurological disorders that exhibit NF-L depolymerization, liberation from neuron-specific compartments, mobilization and enrichment into pathological biofluids; (ii) will evaluate how NF-L exhibits compartmentalization effects in age-related neurological disorders; (iii) will review how the shift of NF-L compartmentalization from within the neuronal cytoskeleton into peripheral biofluids may be a diagnostic biomarker for neuronal-decline in all cause dementia most useful in distinguishing between closely related neurological disorders; and (iv) will review emerging evidence that deficits in plasma membrane barrier integrity, pathological transport and/or vesicle-mediated trafficking dysfunction of NF-L may contribute to neuronal decline, with specific reference to AD wherever possible.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Cell Biology and Anatomy, LSU Health Science Center, New Orleans LA 70112, USA
| | - Lisa Arceneaux
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| | - Frank Culicchia
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Neurosurgery, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans LA 7011, USA
- Department of Neurology, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| |
Collapse
|
32
|
Graham NSN, Zimmerman KA, Moro F, Heslegrave A, Maillard SA, Bernini A, Miroz JP, Donat CK, Lopez MY, Bourke N, Jolly AE, Mallas EJ, Soreq E, Wilson MH, Fatania G, Roi D, Patel MC, Garbero E, Nattino G, Baciu C, Fainardi E, Chieregato A, Gradisek P, Magnoni S, Oddo M, Zetterberg H, Bertolini G, Sharp DJ. Axonal marker neurofilament light predicts long-term outcomes and progressive neurodegeneration after traumatic brain injury. Sci Transl Med 2021; 13:eabg9922. [PMID: 34586833 DOI: 10.1126/scitranslmed.abg9922] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Neil S N Graham
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Karl A Zimmerman
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Federico Moro
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo 24126, Italy.,Fondazione IRCCS, Ca' Granda Ospedale Maggiore Policlinico, Dipartimento di Anestesia e Rianimazione, 20122, Milan, Italy
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.,UK Dementia Research Institute at UCL, London WC1N 3BG, UK
| | - Samia Abed Maillard
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Adriano Bernini
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - John-Paul Miroz
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Maria Yanez Lopez
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Niall Bourke
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Amy E Jolly
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Emma-Jane Mallas
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Eyal Soreq
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK
| | - Mark H Wilson
- Department of Neurosurgery, Imperial College Healthcare NHS Trust, London W6 8RF, UK.,Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Gavin Fatania
- Department of Imaging, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Dylan Roi
- Department of Imaging, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Maneesh C Patel
- Department of Imaging, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Elena Garbero
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo 24126, Italy
| | - Giovanni Nattino
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo 24126, Italy
| | - Camelia Baciu
- Neurorianimazione, ASST Grande Ospedale Metropolitano Niguarda, Milano 20162, Italy
| | - Enrico Fainardi
- Department of Experimental and Clinical Sciences, Careggi University Hospital, University of Firenze, Florence 50139, Italy
| | - Arturo Chieregato
- Neurorianimazione, ASST Grande Ospedale Metropolitano Niguarda, Milano 20162, Italy
| | - Primoz Gradisek
- Clinical Department of Anaesthesiology and Intensive Therapy, University Medical Center, Ljubljana 1000, Slovenia
| | - Sandra Magnoni
- Department of Anesthesia and Intensive Care, Santa Chiara Hospital, Trento 38122, Italy
| | - Mauro Oddo
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland.,Medical Direction, CHUV Lausanne University Hospital, Lausanne 1011, Switzerland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal 431 41, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 431 41, Sweden
| | - Guido Bertolini
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo 24126, Italy
| | - David J Sharp
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK DRI Centre for Care Research and Technology, Imperial College London, London W12 0BZ, UK.,Centre for Injury Studies, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
33
|
18F-florbetapir PET as a marker of myelin integrity across the Alzheimer's disease spectrum. Eur J Nucl Med Mol Imaging 2021; 49:1242-1253. [PMID: 34581847 PMCID: PMC8921113 DOI: 10.1007/s00259-021-05493-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/08/2021] [Indexed: 01/23/2023]
Abstract
Purpose Recent evidence suggests that PET imaging with amyloid-β (Aβ) tracers can be used to assess myelin integrity in cerebral white matter (WM). Alzheimer’s disease (AD) is characterized by myelin changes that are believed to occur early in the disease course. Nevertheless, the extent to which demyelination, as measured with Aβ PET, contributes to AD progression remains unexplored. Methods Participants with concurrent 18F-florbetapir (FBP) PET, MRI, and cerebrospinal fluid (CSF) examinations were included (241 cognitively normal, 347 Aβ-positive cognitively impaired, and 207 Aβ-negative cognitively impaired subjects). A subset of these participants had also available diffusion tensor imaging (DTI) images (n = 195). We investigated cross-sectional associations of FBP retention in the white matter (WM) with MRI-based markers of WM degeneration, AD clinical progression, and fluid biomarkers. In longitudinal analyses, we used linear mixed models to assess whether FBP retention in normal-appearing WM (NAWM) predicted progression of WM hyperintensity (WMH) burden and clinical decline. Results In AD-continuum individuals, FBP retention in NAWM was (1) higher compared with WMH regions, (2) associated with DTI-based measures of WM integrity, and (3) associated with longitudinal progression of WMH burden. FBP uptake in WM decreased across the AD continuum and with increasingly abnormal CSF biomarkers of AD. Furthermore, FBP retention in the WM was associated with large-calibre axon degeneration as reflected by abnormal plasma neurofilament light chain levels. Low FBP uptake in NAWM predicted clinical decline in preclinical and prodromal AD, independent of demographics, global cortical Aβ, and WMH burden. Most of these associations were also observed in Aβ-negative cognitively impaired individuals. Conclusion These results support the hypothesis that FBP retention in the WM is myelin-related. Demyelination levels progressed across the AD continuum and were associated with clinical progression at early stages, suggesting that this pathologic process might be a relevant degenerative feature in the disease course. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05493-y.
Collapse
|
34
|
Yuan A, Nixon RA. Neurofilament Proteins as Biomarkers to Monitor Neurological Diseases and the Efficacy of Therapies. Front Neurosci 2021; 15:689938. [PMID: 34646114 PMCID: PMC8503617 DOI: 10.3389/fnins.2021.689938] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Biomarkers of neurodegeneration and neuronal injury have the potential to improve diagnostic accuracy, disease monitoring, prognosis, and measure treatment efficacy. Neurofilament proteins (NfPs) are well suited as biomarkers in these contexts because they are major neuron-specific components that maintain structural integrity and are sensitive to neurodegeneration and neuronal injury across a wide range of neurologic diseases. Low levels of NfPs are constantly released from neurons into the extracellular space and ultimately reach the cerebrospinal fluid (CSF) and blood under physiological conditions throughout normal brain development, maturation, and aging. NfP levels in CSF and blood rise above normal in response to neuronal injury and neurodegeneration independently of cause. NfPs in CSF measured by lumbar puncture are about 40-fold more concentrated than in blood in healthy individuals. New ultra-sensitive methods now allow minimally invasive measurement of these low levels of NfPs in serum or plasma to track disease onset and progression in neurological disorders or nervous system injury and assess responses to therapeutic interventions. Any of the five Nf subunits - neurofilament light chain (NfL), neurofilament medium chain (NfM), neurofilament heavy chain (NfH), alpha-internexin (INA) and peripherin (PRPH) may be altered in a given neuropathological condition. In familial and sporadic Alzheimer's disease (AD), plasma NfL levels may rise as early as 22 years before clinical onset in familial AD and 10 years before sporadic AD. The major determinants of elevated levels of NfPs and degradation fragments in CSF and blood are the magnitude of damaged or degenerating axons of fiber tracks, the affected axon caliber sizes and the rate of release of NfP and fragments at different stages of a given neurological disease or condition directly or indirectly affecting central nervous system (CNS) and/or peripheral nervous system (PNS). NfPs are rapidly emerging as transformative blood biomarkers in neurology providing novel insights into a wide range of neurological diseases and advancing clinical trials. Here we summarize the current understanding of intracellular NfP physiology, pathophysiology and extracellular kinetics of NfPs in biofluids and review the value and limitations of NfPs and degradation fragments as biomarkers of neurodegeneration and neuronal injury.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Psychiatry, NYU Neuroscience Institute, New York, NY, United States
- Department of Cell Biology, New York University Grossman School of Medicine, (NYU), Neuroscience Institute, New York, NY, United States
| |
Collapse
|
35
|
Li D, Zhang L, Nelson NW, Mielke MM, Yu F. Plasma Neurofilament Light and Future Declines in Cognition and Function in Alzheimer's Disease in the FIT-AD Trial. J Alzheimers Dis Rep 2021; 5:601-611. [PMID: 34514342 PMCID: PMC8385429 DOI: 10.3233/adr-210302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2021] [Indexed: 01/27/2023] Open
Abstract
Background: Utilities of blood-based biomarkers in Alzheimer’s disease (AD) clinical trials remain unknown. Objective: To evaluate the ability of plasma neurofilament light chain (NfL) to predict future declines in cognition and activities of daily living (ADL) outcomes in 26 older adults with mild-to-moderate AD dementia from the FIT-AD Trial. Methods: Plasma NfL was measured at baseline and 3 and 6 months. Cognition and ADL were assessed using the AD Assessment Scale-Cognition (ADAS-Cog) and AD Uniform Dataset Instruments and Disability Assessment for Dementia (DAD), respectively, at baseline, 3, 6, 9, and 12 months. Linear mixed effects models were used to examine the associations between baseline or change in plasma NfL and changes in outcomes. Results: Higher baseline plasma NfL was associated with greater rate of decline in ADAS-Cog from baseline to 6 months (standardized estimate of 0.00462, p = 0.02853) and in ADL from baseline to 12 months (standardized estimate of –0.00284, p = 0.03338). Greater increase in plasma NfL in short term from baseline to 3 months was associated with greater rate of decline in memory and ADL from 3 to 6 months (standardized estimate of –0.04638 [0.003], p = 0.01635; standardized estimate of –0.03818, p = 0.0435) and greater rate of decline in ADL from 3 to 12 month (standardized estimate of –0.01492, p = 0.01082). Conclusion: This study demonstrated that plasma NfL might have the potential to predict cognitive and function decline up to 12 months. However, future studies with bigger sample sizes need to confirm the findings.
Collapse
Affiliation(s)
- Danni Li
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Lin Zhang
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Nathaniel W Nelson
- Graduate School of Professional Psychology, University of St. Thomas, Minnesota, St. Paul, MN, USA
| | - Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Fang Yu
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| |
Collapse
|
36
|
He L, de Souto Barreto P, Giudici KV, Aggarwal G, Nguyen AD, Morley JE, Li Y, Bateman RJ, Vellas B. Cross-Sectional and Longitudinal Associations Between Plasma Neurodegenerative Biomarkers and Physical Performance Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2021; 76:1874-1881. [PMID: 33186456 DOI: 10.1093/gerona/glaa284] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Plasma amyloid-beta (Aβ), neurofilament light chain (NfL), and progranulin (PGRN) have been related to multiple neurodegenerative conditions that might affect physical performance. The aim of this study was to explore the relationship between these plasma neurodegenerative markers and physical performance among community-dwelling older adults. METHODS Five hundred and seven older adults (aged 76 ± 5 years) previously recruited in the Multidomain Alzheimer's Preventive Trial, and had received blood and physical performance tests, were included in this study. Plasma Aβ (Aβ 42/Aβ 40 ratio), NfL, and PGRN levels were measured. Physical performance was assessed by handgrip strength and the Short Physical Performance Battery (combining gait speed, chair stands, and balance tests). Physical performance measured at the same time point and after the blood tests were used. Mixed-effect linear models were performed with age, sex, allocation to Multidomain Alzheimer's Preventive Trial group, body mass index, and Mini-Mental State Examination score as covariates. RESULTS The mean values of Aβ 42/Aβ 40 ratio, NfL, and PGRN were 0.11, 84.06 pg/mL, and 45.43 ng/mL, respectively. At the cross-sectional level, higher plasma NfL was associated with a lower Short Physical Performance Battery score (β = -0.004, 95% CI [-0.007, -0.001]). At the longitudinal level, higher PGRN levels were associated with decreasing handgrip strength over time (β = -0.02, 95% CI [-0.04, -0.007]). All the other associations were statistically nonsignificant. CONCLUSION Our findings suggest the possibility of using plasma NfL and PGRN as markers of physical performance in older adults.
Collapse
Affiliation(s)
- Lingxiao He
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France.,UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Kelly V Giudici
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Missouri
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Missouri
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France.,UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | | |
Collapse
|
37
|
Alagaratnam J, Stöhr W, Toombs J, Heslegrave A, Zetterberg H, Gisslén M, Pett S, Nelson M, Clarke A, Nwokolo N, Johnson MA, Khan M, Hanke T, Kopycinski J, Dorrell L, Fox J, Kinloch S, Underwood J, Pace M, Frater J, Winston A, Fidler S. No evidence of neuronal damage as measured by neurofilament light chain in a HIV cure study utilising a kick-and-kill approach. J Virus Erad 2021; 7:100056. [PMID: 34611495 PMCID: PMC8477217 DOI: 10.1016/j.jve.2021.100056] [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: 11/29/2020] [Revised: 08/03/2021] [Accepted: 09/08/2021] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE HIV-remission strategies including kick-and-kill could induce viral transcription and immune-activation in the central nervous system, potentially causing neuronal injury. We investigated the impact of kick-and-kill on plasma neurofilament light (NfL), a marker of neuro-axonal injury, in RIVER trial participants commencing antiretroviral treatment (ART) during primary infection and randomly allocated to ART-alone or kick-and-kill (ART + vaccination + vorinostat (ART + V + V)). DESIGN Sub-study measuring serial plasma NfL concentrations. METHODS Plasma NfL (using Simoa digital immunoassay), plasma HIV-1 RNA (using single-copy assay) and total HIV-1 DNA (using quantitative polymerase chain reaction in peripheral CD4+ T-cells) were measured at randomisation (following ≥22 weeks ART), week 12 (on final intervention day in ART + V + V) and week 18 post-randomisation. HIV-specific T-cells were quantified by intracellular cytokine staining at randomisation and week 12. Differences in plasma NfL longitudinally and by study arm were analysed using mixed models and Student's t-test. Associations with plasma NfL were assessed using linear regression and rank statistics. RESULTS At randomisation, 58 male participants had median age 32 years and CD4+ count 696 cells/μL. No significant difference in plasma NfL was seen longitudinally and by study arm, with median plasma NfL (pg/mL) in ART-only vs ART + V + V: 7.4 vs 6.4, p = 0.16 (randomisation), 8.0 vs 6.9, p = 0.22 (week 12) and 7.1 vs 6.8, p = 0.74 (week 18). Plasma NfL did not significantly correlate with plasma HIV-1 RNA and total HIV-1 DNA concentration in peripheral CD4+ T-cells at any timepoint. While higher HIV-specific T-cell responses were seen at week 12 in ART + V + V, there were no significant correlations with plasma NfL. In multivariate analysis, higher plasma NfL was associated with older age, higher CD8+ count and lower body mass index. CONCLUSIONS Despite evidence of vaccine-induced HIV-specific T-cell responses, we observed no evidence of increased neuro-axonal injury using plasma NfL as a biomarker up to 18 weeks following kick-and-kill, compared with ART-only.
Collapse
Affiliation(s)
- Jasmini Alagaratnam
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
- Genitourinary Medicine and HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, W2 1NY, United Kingdom
| | - Wolfgang Stöhr
- Medical Research Council Clinical Trials Unit at UCL, 90 High Holborn, Holborn, London, WC1V 6LJ, United Kingdom
| | - Jamie Toombs
- UK Dementia Research Institute at University College London, UCL Cruciform Building, Gower Street, Bloomsbury, London, WC1E 6BT, UK
| | - Amanda Heslegrave
- UK Dementia Research Institute at University College London, UCL Cruciform Building, Gower Street, Bloomsbury, London, WC1E 6BT, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Cruciform Building, Gower Street, Bloomsbury, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Wallingsgatan 6, 431 41, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 5, 413 45, Göteborg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Blå Stråket 5, 413 45, Göteborg, Sweden
| | - Sarah Pett
- Medical Research Council Clinical Trials Unit at UCL, 90 High Holborn, Holborn, London, WC1V 6LJ, United Kingdom
- Institute for Global Health, University College London, Gower St, Bloomsbury, London, WC1E 6BT, UK
- Mortimer Market Centre, Central and North West London NHS Foundation Trust, Capper St, Bloomsbury, London, WC1E 6JB, UK
| | - Mark Nelson
- Department of Genitourinary Medicine and HIV, Chelsea & Westminster NHS Foundation Trust, 369 Fulham Rd, Chelsea, London, SW10 9NH, UK
| | - Amanda Clarke
- Department of Genitourinary Medicine and HIV, Brighton & Sussex University Hospitals NHS Trust, Kemptown, Brighton, BN2 1ES, UK
| | - Nneka Nwokolo
- Department of Genitourinary Medicine and HIV, Chelsea & Westminster NHS Foundation Trust, 369 Fulham Rd, Chelsea, London, SW10 9NH, UK
| | - Margaret A. Johnson
- Department of Infection and Immunity, Royal Free Hospital, Pond Street, London, NW3 2QG, United Kingdom
| | - Maryam Khan
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
| | - Tomas Hanke
- The Jenner Institute, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, OX3 7DQ, UK
- The Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| | - Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Lucy Dorrell
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Julie Fox
- Department of Genitourinary Medicine and HIV, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, SE1 9RT, UK
| | - Sabine Kinloch
- Department of Infection and Immunity, Royal Free Hospital, Pond Street, London, NW3 2QG, United Kingdom
| | - Jonathan Underwood
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
- Division of Infection and Immunity, School of Medicine, Cardiff University, School of Medicine, UHW Main Building, Heath Park, Cardiff, CF14 4XN, UK
| | - Matthew Pace
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, South Parks Road, Oxford, OX1 3SY, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, South Parks Road, Oxford, OX1 3SY, UK
- Oxford University National Institute of Health Research Biomedical Research Centre, Oxford, OX1 2JD, UK
| | - Alan Winston
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
- Genitourinary Medicine and HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, W2 1NY, United Kingdom
| | - Sarah Fidler
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
- Genitourinary Medicine and HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, W2 1NY, United Kingdom
| | - the RIVER trial study group
- Department of Infectious Disease, St Mary's Hospital Campus, Imperial College London, London, W2 1NY, United Kingdom
- Genitourinary Medicine and HIV Department, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, W2 1NY, United Kingdom
- Medical Research Council Clinical Trials Unit at UCL, 90 High Holborn, Holborn, London, WC1V 6LJ, United Kingdom
- UK Dementia Research Institute at University College London, UCL Cruciform Building, Gower Street, Bloomsbury, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Wallingsgatan 6, 431 41, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 5, 413 45, Göteborg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Blå Stråket 5, 413 45, Göteborg, Sweden
- Institute for Global Health, University College London, Gower St, Bloomsbury, London, WC1E 6BT, UK
- Mortimer Market Centre, Central and North West London NHS Foundation Trust, Capper St, Bloomsbury, London, WC1E 6JB, UK
- Department of Genitourinary Medicine and HIV, Chelsea & Westminster NHS Foundation Trust, 369 Fulham Rd, Chelsea, London, SW10 9NH, UK
- Department of Genitourinary Medicine and HIV, Brighton & Sussex University Hospitals NHS Trust, Kemptown, Brighton, BN2 1ES, UK
- Department of Infection and Immunity, Royal Free Hospital, Pond Street, London, NW3 2QG, United Kingdom
- The Jenner Institute, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, OX3 7DQ, UK
- The Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
- Nuffield Department of Medicine, University of Oxford, Oxford, OX1 2JD, UK
- Department of Genitourinary Medicine and HIV, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, SE1 9RT, UK
- Division of Infection and Immunity, School of Medicine, Cardiff University, School of Medicine, UHW Main Building, Heath Park, Cardiff, CF14 4XN, UK
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, South Parks Road, Oxford, OX1 3SY, UK
- Oxford University National Institute of Health Research Biomedical Research Centre, Oxford, OX1 2JD, UK
| |
Collapse
|
38
|
Beydoun MA, Noren Hooten N, Beydoun HA, Maldonado AI, Weiss J, Evans MK, Zonderman AB. Plasma neurofilament light as a potential biomarker for cognitive decline in a longitudinal study of middle-aged urban adults. Transl Psychiatry 2021; 11:436. [PMID: 34420032 PMCID: PMC8380245 DOI: 10.1038/s41398-021-01563-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Plasma neurofilament light (NfL) is a marker for neurodegenerative diseases. Few studies have examined the association of NfL with middle-aged changes in cognitive performance, and no studies have examined differential NfL effects by race. Using data from the Healthy Aging in Neighborhoods of Diversity across the Life Span (HANDLS) study (n = 625, Agev1: 30-66 y, 41.6% male, 56.3% African American, 27.8% below poverty), we investigated the associations of initial NfL levels and annualized change with cognitive performance over time in global mental status, verbal and visual memory, fluency, attention, and executive function. We used ordinary least squares and mixed-effects regressions stratified by race, while exploring differential associations by age group, sex, and poverty status. Over a mean follow-up of 4.3 years, we found initial NfL level was associated with a faster decline on normalized mental status scores in Whites only and in those >50 years old. Annualized increase in NfL was associated with a greater decline in verbal fluency in men. In other exploratory analyses, annualized increase in NfL was associated with a slower decline in verbal memory among individuals living above poverty; in the older group (>50 years), first-visit NfL was linked with better performance at baseline in global mental status and verbal memory. In summary, first-visit NfL was primarily associated with the global mental status decline among Whites, while exhibiting inconsistent relationships in some exploratory analyses. Plasma NfL levels can be detected and quantified in non-demented middle-aged adults and changes can be analyzed over time. More longitudinal studies are needed to address the clinical utility of this biomarker for early cognitive defects.
Collapse
Affiliation(s)
- May A. Beydoun
- grid.419475.a0000 0000 9372 4913Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD USA
| | - Nicole Noren Hooten
- grid.419475.a0000 0000 9372 4913Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD USA
| | - Hind A. Beydoun
- grid.413661.70000 0004 0595 1323Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA USA
| | - Ana I. Maldonado
- grid.419475.a0000 0000 9372 4913Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD USA ,grid.266673.00000 0001 2177 1144Department of Psychology, University of Maryland, Baltimore County, Catonsville, MD USA
| | - Jordan Weiss
- grid.47840.3f0000 0001 2181 7878Department of Demography, University of California, Berkeley, Berkeley, CA USA
| | - Michele K. Evans
- grid.419475.a0000 0000 9372 4913Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD USA
| | - Alan B. Zonderman
- grid.419475.a0000 0000 9372 4913Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD USA
| |
Collapse
|
39
|
Circulating Biomarkers in Neuromuscular Disorders: What Is Known, What Is New. Biomolecules 2021; 11:biom11081246. [PMID: 34439911 PMCID: PMC8393752 DOI: 10.3390/biom11081246] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
The urgent need for new therapies for some devastating neuromuscular diseases (NMDs), such as Duchenne muscular dystrophy or amyotrophic lateral sclerosis, has led to an intense search for new potential biomarkers. Biomarkers can be classified based on their clinical value into different categories: diagnostic biomarkers confirm the presence of a specific disease, prognostic biomarkers provide information about disease course, and therapeutic biomarkers are designed to predict or measure treatment response. Circulating biomarkers, as opposed to instrumental/invasive ones (e.g., muscle MRI or nerve ultrasound, muscle or nerve biopsy), are generally easier to access and less “time-consuming”. In addition to well-known creatine kinase, other promising molecules seem to be candidate biomarkers to improve the diagnosis, prognosis and prediction of therapeutic response, such as antibodies, neurofilaments, and microRNAs. However, there are some criticalities that can complicate their application: variability during the day, stability, and reliable performance metrics (e.g., accuracy, precision and reproducibility) across laboratories. In the present review, we discuss the application of biochemical biomarkers (both validated and emerging) in the most common NMDs with a focus on their diagnostic, prognostic/predictive and therapeutic application, and finally, we address the critical issues in the introduction of new biomarkers.
Collapse
|
40
|
Merkulyeva N, Mikhalkin A. SMI-32 labeling in Cajal-Retzius cells of feline primary visual cortex. Neurosci Lett 2021; 762:136165. [PMID: 34371123 DOI: 10.1016/j.neulet.2021.136165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022]
Abstract
Cajal-Retzius cells are one of the transient elements of the developing cerebral cortex. These cells express some characteristic molecules. One of them, heavy-chain neurofilaments, participating in the construction of the mature cerebral networks, are believed to be a specific feature of the human's Cajal-Retzius cells. Using histochemical stain for SMI-32 antibody to the non-phosphorylated heavy-chain neurofilaments, large neurons having horizontally oriented soma and bipolar processes were labeled in the molecular layer of the primary visual cortex of cats aged 0-2 postnatal days. Using DiI technique, similar neurons having a well-developed system of parallel vertical branches coming from the two horizontal processes were visualized in these areas. The location and general morphology of these neurons were similar to the Cajal-Retzius cells allowing to suppose for the carnivores to share similar with primates developmental mechanisms of the corticogenesis.
Collapse
Affiliation(s)
- Natalia Merkulyeva
- Lab Neuromorphology, Pavlov Institute of Physiology RAS Russia, Saint-Petersburg, Makarov emb., 6, 199034, Russia.
| | - Aleksandr Mikhalkin
- Lab Neuromorphology, Pavlov Institute of Physiology RAS Russia, Saint-Petersburg, Makarov emb., 6, 199034, Russia
| |
Collapse
|
41
|
Mages B, Fuhs T, Aleithe S, Blietz A, Hobusch C, Härtig W, Schob S, Krueger M, Michalski D. The Cytoskeletal Elements MAP2 and NF-L Show Substantial Alterations in Different Stroke Models While Elevated Serum Levels Highlight Especially MAP2 as a Sensitive Biomarker in Stroke Patients. Mol Neurobiol 2021; 58:4051-4069. [PMID: 33931805 PMCID: PMC8280005 DOI: 10.1007/s12035-021-02372-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
In the setting of ischemic stroke, the neurofilament subunit NF-L and the microtubule-associated protein MAP2 have proven to be exceptionally ischemia-sensitive elements of the neuronal cytoskeleton. Since alterations of the cytoskeleton have been linked to the transition from reversible to irreversible tissue damage, the present study investigates underlying time- and region-specific alterations of NF-L and MAP2 in different animal models of focal cerebral ischemia. Although NF-L is increasingly established as a clinical stroke biomarker, MAP2 serum measurements after stroke are still lacking. Therefore, the present study further compares serum levels of MAP2 with NF-L in stroke patients. In the applied animal models, MAP2-related immunofluorescence intensities were decreased in ischemic areas, whereas the abundance of NF-L degradation products accounted for an increase of NF-L-related immunofluorescence intensity. Accordingly, Western blot analyses of ischemic areas revealed decreased protein levels of both MAP2 and NF-L. The cytoskeletal alterations are further reflected at an ultrastructural level as indicated by a significant reduction of detectable neurofilaments in cortical axons of ischemia-affected areas. Moreover, atomic force microscopy measurements confirmed altered mechanical properties as indicated by a decreased elastic strength in ischemia-affected tissue. In addition to the results from the animal models, stroke patients exhibited significantly elevated serum levels of MAP2, which increased with infarct size, whereas serum levels of NF-L did not differ significantly. Thus, MAP2 appears to be a more sensitive stroke biomarker than NF-L, especially for early neuronal damage. This perspective is strengthened by the results from the animal models, showing MAP2-related alterations at earlier time points compared to NF-L. The profound ischemia-induced alterations further qualify both cytoskeletal elements as promising targets for neuroprotective therapies.
Collapse
Affiliation(s)
- Bianca Mages
- Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Thomas Fuhs
- Section of Soft Matter Physics, Faculty of Physics and Geosciences, Leipzig University, Leipzig, Germany
| | - Susanne Aleithe
- Department of Neurology, Leipzig University, Leipzig, Germany
| | | | | | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Stefan Schob
- Department of Neuroradiology, Leipzig University, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
42
|
Garrudo FFF, Nogueira DES, Rodrigues CAV, Ferreira FA, Paradiso P, Colaço R, Marques AC, Cabral JMS, Morgado J, Linhardt RJ, Ferreira FC. Electrical stimulation of neural-differentiating iPSCs on novel coaxial electroconductive nanofibers. Biomater Sci 2021; 9:5359-5382. [PMID: 34223566 DOI: 10.1039/d1bm00503k] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neural tissue engineering strategies are paramount to create fully mature neurons, necessary for new therapeutic strategies for neurological diseases or the creation of reliable in vitro models. Scaffolds can provide physical support for these neurons and enable cues for enhancing neural cell differentiation, such as electrical current. Coaxial electrospinning fibers, designed to fulfill neural cell needs, bring together an electroconductive shell layer (PCL-PANI), able to mediate electrical stimulation of cells cultivated on fibers mesh surface, and a soft core layer (PGS), used to finetune fiber diameter (951 ± 465 nm) and mechanical properties (1.3 ± 0.2 MPa). Those dual functional coaxial fibers are electroconductive (0.063 ± 0.029 S cm-1, stable over 21 days) and biodegradable (72% weigh loss in 12 hours upon human lipase accelerated assay). For the first time, the long-term effects of electrical stimulation on induced neural progenitor cells were studied using such fibers. The results show increase in neural maturation (upregulation of MAP2, NEF-H and SYP), up-regulation of glutamatergic marker genes (VGLUT1 - 15-fold) and voltage-sensitive channels (SCN1α - 12-fold, CACNA1C - 32-fold), and a down-regulation of GABAergic marker (GAD67 - 0.09-fold), as detected by qRT-PCR. Therefore, this study suggest a shift from an inhibitory to an excitatory neural cell profile. This work shows that the PGS/PCL-PANI coaxial fibers here developed have potential applications in neural tissue engineering.
Collapse
Affiliation(s)
- Fábio F F Garrudo
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA. and Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal and Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Diogo E S Nogueira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Flávio A Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Patrizia Paradiso
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Rogério Colaço
- IDMEC - Instituto de Engenharia Mecânica, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Ana C Marques
- CERENA, DEQ, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, P-1049-001 Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Jorge Morgado
- Department of Bioengineering and Instituto de Telecomunicações, Universidade de Lisboa, Av. Rovisco Pais, P-1049-001, Lisboa, Portugal
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Department of Chemistry & Chemical Biology, Rensselaer Polytechnic Institute, Biotechnology Center 4005, Troy, NY 12180, USA.
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal. and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
43
|
Jakob MO, Kofoed-Branzk M, Deshpande D, Murugan S, Klose CSN. An Integrated View on Neuronal Subsets in the Peripheral Nervous System and Their Role in Immunoregulation. Front Immunol 2021; 12:679055. [PMID: 34322118 PMCID: PMC8312561 DOI: 10.3389/fimmu.2021.679055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
The peripheral nervous system consists of sensory circuits that respond to external and internal stimuli and effector circuits that adapt physiologic functions to environmental challenges. Identifying neurotransmitters and neuropeptides and the corresponding receptors on immune cells implies an essential role for the nervous system in regulating immune reactions. Vice versa, neurons express functional cytokine receptors to respond to inflammatory signals directly. Recent advances in single-cell and single-nuclei sequencing have provided an unprecedented depth in neuronal analysis and allowed to refine the classification of distinct neuronal subsets of the peripheral nervous system. Delineating the sensory and immunoregulatory capacity of different neuronal subsets could inform a better understanding of the response happening in tissues that coordinate physiologic functions, tissue homeostasis and immunity. Here, we summarize current subsets of peripheral neurons and discuss neuronal regulation of immune responses, focusing on neuro-immune interactions in the gastrointestinal tract. The nervous system as a central coordinator of immune reactions and tissue homeostasis may predispose for novel promising therapeutic approaches for a large variety of diseases including but not limited to chronic inflammation.
Collapse
Affiliation(s)
- Manuel O Jakob
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Kofoed-Branzk
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Divija Deshpande
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Shaira Murugan
- Department of BioMedical Research, Group of Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
44
|
Neto WK, Gama EF, de Assis Silva W, de Oliveira TVA, Dos Santos Vilas Boas AE, Ciena AP, Anaruma CA, Caperuto ÉC. Ladder-based resistance training elicited similar ultrastructural adjustments in forelimb and hindlimb peripheral nerves of young adult Wistar rats. Exp Brain Res 2021; 239:2583-2592. [PMID: 34191117 DOI: 10.1007/s00221-021-06156-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/19/2021] [Indexed: 12/22/2022]
Abstract
To analyze the morphological response induced by high-volume, high-intensity ladder-based resistance training (LRT) on the ultrastructure of the radial (forelimb) and sciatic (hindlimb) nerves of adults Wistar rats. Twenty rats were equally distributed into groups: sedentary (SED) and LRT. After the rodents were subjected to the maximum load (ML) carrying test, the LRT group performed 6-8 progressive climbs (2 × 50% ML, 2 × 75% ML, 2 × 100% ML, and 2 × 100% ML + 30 g) three times per week. After 8 weeks, the radial and sciatic nerves were removed and prepared for transmission electron microscopy. In the radial nerve, myelinated axons cross-sectional area (CSA), unmyelinated axons CSA, myelin sheath thickness, and Schwann cells nuclei area were statistically larger in the LRT group than SED (p < 0.05). Also, the number of microtubules and neurofilaments per field were statistically higher in the LRT group than in SED (p < 0.01). For sciatic nerve, myelinated fibers CSA, unmyelinated axons CSA, myelin sheath thickness, Schwann cells nuclei area, and the number of neurofilaments per field were statistically larger in the LRT group compared to the SED group (p < 0.05). LRT with high-volume and high-intensity effectively induce similar changes in adult Wistar rats' radial and sciatic nerves' ultrastructure.
Collapse
Affiliation(s)
- Walter Krause Neto
- Department of Physical Education, Laboratory of Morphoquantitative Studies and Immunohistochemistry, São Judas Tadeu University, Rua Taquari, 546-Mooca Unit, P. O. Box 03166-000, São Paulo, SP, Brazil.
| | - Eliane Florencio Gama
- Department of Morphology, Faculty of Medical Sciences, Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Wellington de Assis Silva
- Department of Physical Education, Laboratory of Morphoquantitative Studies and Immunohistochemistry, São Judas Tadeu University, Rua Taquari, 546-Mooca Unit, P. O. Box 03166-000, São Paulo, SP, Brazil
| | - Tony Vinicius Apolinário de Oliveira
- Department of Physical Education, Laboratory of Morphoquantitative Studies and Immunohistochemistry, São Judas Tadeu University, Rua Taquari, 546-Mooca Unit, P. O. Box 03166-000, São Paulo, SP, Brazil
| | - Alan Esaú Dos Santos Vilas Boas
- Department of Physical Education, Laboratory of Morphoquantitative Studies and Immunohistochemistry, São Judas Tadeu University, Rua Taquari, 546-Mooca Unit, P. O. Box 03166-000, São Paulo, SP, Brazil
| | - Adriano Polican Ciena
- Department of Physical Education, Laboratory of Morphology and Physical Activity, São Paulo State University "Júlio de Mesquita Filho", Rio Claro, SP, Brazil
| | - Carlos Alberto Anaruma
- Department of Physical Education, Laboratory of Morphology and Physical Activity, São Paulo State University "Júlio de Mesquita Filho", Rio Claro, SP, Brazil
| | - Érico Chagas Caperuto
- Depatment of Physical Education, Laboratory of Human Movement, São Judas Tadeu University, São Paulo, SP, Brazil
| |
Collapse
|
45
|
He L, Morley JE, Aggarwal G, Nguyen AD, Vellas B, de Souto Barreto P. Plasma neurofilament light chain is associated with cognitive decline in non-dementia older adults. Sci Rep 2021; 11:13394. [PMID: 34183688 PMCID: PMC8238930 DOI: 10.1038/s41598-021-91038-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023] Open
Abstract
Neurofilament light chain (NfL) has been associated with cognitive status in multiple neurodegenerative conditions. Studies about plasma NfL and cognitive decline in older adults are still limited. 504 older adults (median age 75 years) who expressed memory complaints were selected from the Multidomain Alzheimer's Preventive Trial (MAPT) and were classified as normal cognition (NC) or mild cognitive impairment (MCI). Cognitive functions were measured as mini mental state examination (MMSE) and composite cognitive score (CCS) over a 4-year period. Plasma NfL was measured at the first or the second year of the MAPT. Mixed-effects linear models were performed to evaluate cross-sectional and longitudinal associations. In the whole population, higher plasma NfL was cross-sectionally associated with lower cognitive functions (MMSE: β = - 0.007, 95% CI [- 0.013, - 0.001]; CCS: β = - 0.003, 95% CI [- 0.006, - 0.001]). In adults with MCI, but not NC, higher plasma NfL was associated with lower CCS at the cross-sectional level (β = - 0.003, 95% CI [- 0.005, - 0.0002]). The upper quartile NfL group further demonstrated more over time decline in CCS (β = - 0.07, 95% CI [- 0.12, - 0.01]) under the MCI status. Plasma NfL can be a promising biomarker of progressive cognition decline in older adults with MCI.
Collapse
Affiliation(s)
- Lingxiao He
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, 37 Allées Jules Guesdes, 31000, Toulouse, France.
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, USA
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, 37 Allées Jules Guesdes, 31000, Toulouse, France
- CERPOP, Inserm 1295, Université de Toulouse, UPS, 31000, Toulouse, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, 37 Allées Jules Guesdes, 31000, Toulouse, France
- CERPOP, Inserm 1295, Université de Toulouse, UPS, 31000, Toulouse, France
| |
Collapse
|
46
|
Bergström S, Remnestål J, Yousef J, Olofsson J, Markaki I, Carvalho S, Corvol JC, Kultima K, Kilander L, Löwenmark M, Ingelsson M, Blennow K, Zetterberg H, Nellgård B, Brosseron F, Heneka MT, Bosch B, Sanchez-Valle R, Månberg A, Svenningsson P, Nilsson P. Multi-cohort profiling reveals elevated CSF levels of brain-enriched proteins in Alzheimer's disease. Ann Clin Transl Neurol 2021; 8:1456-1470. [PMID: 34129723 PMCID: PMC8283172 DOI: 10.1002/acn3.51402] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/30/2021] [Accepted: 05/12/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Decreased amyloid beta (Aβ) 42 together with increased tau and phospho-tau in cerebrospinal fluid (CSF) is indicative of Alzheimer's disease (AD). However, the molecular pathophysiology underlying the slowly progressive cognitive decline observed in AD is not fully understood and it is not known what other CSF biomarkers may be altered in early disease stages. METHODS We utilized an antibody-based suspension bead array to analyze levels of 216 proteins in CSF from AD patients, patients with mild cognitive impairment (MCI), and controls from two independent cohorts collected within the AETIONOMY consortium. Two additional cohorts from Sweden were used for biological verification. RESULTS Six proteins, amphiphysin (AMPH), aquaporin 4 (AQP4), cAMP-regulated phosphoprotein 21 (ARPP21), growth-associated protein 43 (GAP43), neurofilament medium polypeptide (NEFM), and synuclein beta (SNCB) were found at increased levels in CSF from AD patients compared with controls. Next, we used CSF levels of Aβ42 and tau for the stratification of the MCI patients and observed increased levels of AMPH, AQP4, ARPP21, GAP43, and SNCB in the MCI subgroups with abnormal tau levels compared with controls. Further characterization revealed strong to moderate correlations between these five proteins and tau concentrations. INTERPRETATION In conclusion, we report six extensively replicated candidate biomarkers with the potential to reflect disease development. Continued evaluation of these proteins will determine to what extent they can aid in the discrimination of MCI patients with and without an underlying AD etiology, and if they have the potential to contribute to a better understanding of the AD continuum.
Collapse
Affiliation(s)
- Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Julia Remnestål
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jamil Yousef
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jennie Olofsson
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stephanie Carvalho
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Assistance-Publique Hôpitaux de Paris, INSERM, CNRS, Hôpital Pitié-Salpêtrière, Department of Neurology, Centre d'Investigation Clinique Neurosciences, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Assistance-Publique Hôpitaux de Paris, INSERM, CNRS, Hôpital Pitié-Salpêtrière, Department of Neurology, Centre d'Investigation Clinique Neurosciences, Paris, France
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Malin Löwenmark
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg.,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
| | - Bengt Nellgård
- Anesthesiology and Intensive Care Medicine, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg
| | - Frederic Brosseron
- Universitätsklinikum Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Beatriz Bosch
- Alzheimer's and other cognitive disorders Unit. Service of Neurology, Hospital Clínic de Barcelona, Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's and other cognitive disorders Unit. Service of Neurology, Hospital Clínic de Barcelona, Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
47
|
Aruta F, Severi D, Iovino A, Spina E, Barghigiani M, Ruggiero L, Iodice R, Santorelli FM, Manganelli F, Tozza S. Proximal weakness involvement in the first Italian case of Charcot-Marie-Tooth 2CC harboring a novel frameshift variant in NEFH. J Peripher Nerv Syst 2021; 26:231-234. [PMID: 33987933 DOI: 10.1111/jns.12454] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Charcot-Marie-Tooth (CMT) diseases are a clinically and genetically heterogeneous group of disorders. Different variants in the neurofilament heavy chain (NEFH) gene have been described to cause the CMT2CC subtype. Here we report the first Italian patient affected by CMT2CC, harboring a novel variant in NEFH. In describing our patient, we also reviewed previously CMT2CC individuals, and suggested to consider NEFH variant if patients have an axonal sensory-motor neuropathy with a prominent proximal muscles involvement with early requirement of walking aids or wheelchair, remembering a motor neuron disorder.
Collapse
Affiliation(s)
- Francesco Aruta
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | - Daniele Severi
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | - Aniello Iovino
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | - Emanuele Spina
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | | | - Lucia Ruggiero
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | - Rosa Iodice
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | | | - Fiore Manganelli
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| | - Stefano Tozza
- Department of Neuroscience, Reproductive and Odontostomatological Science, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
48
|
Mielke MM. Consideration of Sex Differences in the Measurement and Interpretation of Alzheimer Disease-Related Biofluid-Based Biomarkers. J Appl Lab Med 2021; 5:158-169. [PMID: 31811073 DOI: 10.1373/jalm.2019.030023] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The development of cerebrospinal fluid and blood-based biomarkers for Alzheimer disease (AD) and related disorders is rapidly progressing. Such biomarkers may be used clinically to screen the population, to enhance diagnosis, or to help determine prognosis. Although the use of precision medicine methods has contributed to enhanced understanding of the AD pathophysiological changes and development of assays, one aspect not commonly considered is sex differences. CONTENT There are several ways in which sex can affect the concentration or interpretation of biofluid biomarkers. For some markers, concentrations will vary by sex. For others, the concentrations might not vary by sex, but the impact or interpretation may vary by sex depending on the context of use (e.g., diagnostic vs prognostic). Finally, for others, there will be no sex differences in concentrations or their interpretation. This review will first provide a basis for sex differences, including differences in brain structure and function, and the means by which these differences could contribute to sex differences in biomarker concentrations. Next, the current state of sex differences in AD-related biofluid markers (i.e., amyloid-β, phosphorylated τ, total τ, neurofilament light chain, and neurogranin) will be reviewed. Lastly, factors that can lead to the misinterpretation of observed sex differences in biomarkers (either providing evidence for or against) will be considered. SUMMARY This review is intended to provide an impetus to consider sex differences in the measurement and interpretation of AD-related biofluid-based biomarkers.
Collapse
Affiliation(s)
- Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
49
|
Rawal P, Zhao L. Sialometabolism in Brain Health and Alzheimer's Disease. Front Neurosci 2021; 15:648617. [PMID: 33867926 PMCID: PMC8044809 DOI: 10.3389/fnins.2021.648617] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acids refer to a unique family of acidic sugars with a 9-carbon backbone that are mostly found as terminal residues in glycan structures of glycoconjugates including both glycoproteins and glycolipids. The highest levels of sialic acids are expressed in the brain where they regulate neuronal sprouting and plasticity, axon myelination and myelin stability, as well as remodeling of mature neuronal connections. Moreover, sialic acids are the sole ligands for microglial Siglecs (sialic acid-binding immunoglobulin-type lectins), and sialic acid-Siglec interactions have been indicated to play a critical role in the regulation of microglial homeostasis in a healthy brain. The recent discovery of CD33, a microglial Siglec, as a novel genetic risk factor for late-onset Alzheimer's disease (AD), highlights the potential role of sialic acids in the development of microglial dysfunction and neuroinflammation in AD. Apart from microglia, sialic acids have been found to be involved in several other major changes associated with AD. Elevated levels of serum sialic acids have been reported in AD patients. Alterations in ganglioside (major sialic acid carrier) metabolism have been demonstrated as an aggravating factor in the formation of amyloid pathology in AD. Polysialic acids are linear homopolymers of sialic acids and have been implicated to be an important regulator of neurogenesis that contributes to neuronal repair and recovery from neurodegeneration such as in AD. In summary, this article reviews current understanding of neural functions of sialic acids and alterations of sialometabolism in aging and AD brains. Furthermore, we discuss the possibility of looking at sialic acids as a promising novel therapeutic target for AD intervention.
Collapse
Affiliation(s)
- Punam Rawal
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
50
|
Spicer C, Lu CH, Catapano F, Scoto M, Zaharieva I, Malaspina A, Morgan JE, Greensmith L, Muntoni F, Zhou H. The altered expression of neurofilament in mouse models and patients with spinal muscular atrophy. Ann Clin Transl Neurol 2021; 8:866-876. [PMID: 33683023 PMCID: PMC8045929 DOI: 10.1002/acn3.51336] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives To investigate the levels of neurofilaments (NFs) in transgenic mice and patients with spinal muscular atrophy (SMA), and to evaluate their efficacy as a biomarker in SMA. Methods The levels of NF mRNA transcripts were measured by quantitative real‐time PCR in spinal cord from SMA mice. Blood levels of NF heavy chain (NfH) from mice and patients were measured by an in‐house ELISA method. The response of NFs to therapeutic intervention was analysed in severe SMA mice treated with morpholino antisense oligonucleotides. Results Significant changes in NF transcript and protein in spinal cord and protein levels in blood were detected in SMA mice with severe or mild phenotypes, at different time points. A decrease in blood levels of NfH after antisense oligonucleotide treatment was only transient in the mice, despite the persistent benefit on the disease phenotype. A drastic reduction of over 90% in blood levels of NfF was observed in both control and SMA mice during early postnatal development. In contrast, blood levels of NfH were found to be decreased in older SMA children with chronic disease progression. Interpretation Our results show that blood NfH levels are informative in indicating disease onset and response to antisense oligonucleotides treatment in SMA mice, and indicate their potential as a peripheral marker reflecting the pathological status in central nervous system. In older patients with chronic SMA, however, the lower NfH levels may limit their application as biomarker, highlighting the need to continue to pursue additional biomarkers for this group of patients.
Collapse
Affiliation(s)
- Charlotte Spicer
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Ching-Hua Lu
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Neurology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Francesco Catapano
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Irina Zaharieva
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Andrea Malaspina
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Trauma and Neuroscience Centre, Blizard Institute, Barts and The School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jennifer E Morgan
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| | - Linda Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - Haiyan Zhou
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom.,Genetics and Genomic Medicine Teaching and Research Department, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, United Kingdom
| |
Collapse
|