1
|
Schneider TR, Stöckli L, Felbecker A, Nirmalraj PN. Protein fibril aggregation on red blood cells: a potential biomarker to distinguish neurodegenerative diseases from healthy aging. Brain Commun 2024; 6:fcae180. [PMID: 38873003 PMCID: PMC11170662 DOI: 10.1093/braincomms/fcae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 04/19/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
Neurodegenerative diseases like Alzheimer's disease are characterized by the accumulation of misfolded proteins into fibrils in the brain. Atomic force microscopy is a nanoscale imaging technique that can be used to resolve and quantify protein aggregates from oligomers to fibrils. Recently, we characterized protein fibrillar aggregates adsorbed on the surface of red blood cells with atomic force microscopy from patients with neurocognitive disorders, suggesting a novel Alzheimer's disease biomarker. However, the age association of fibril deposits on red blood cells has not yet been studied in detail in healthy adults. Here, we used atomic force microscopy to visualize and quantify fibril coverage on red blood cells in 50 healthy adults and 37 memory clinic patients. Fibrillar protein deposits sporadically appeared in healthy individuals but were much more prevalent in patients with neurodegenerative disease, especially those with Alzheimer's disease as confirmed by positive CSF amyloid beta 1-42/1-40 ratios. The prevalence of fibrils on the red blood cell surface did not significantly correlate with age in either healthy individuals or Alzheimer's disease patients. The overlap in fibril prevalence on red blood cells between Alzheimer's disease and amyloid-negative patients suggests that fibril deposition on red blood cells could occur in various neurodegenerative diseases. Quantifying red blood cell protein fibril morphology and prevalence on red blood cells could serve as a sensitive biomarker for neurodegeneration, distinguishing between healthy individuals and those with neurodegenerative diseases. Future studies that combine atomic force microscopy with immunofluorescence techniques in larger-scale studies could further identify the chemical nature of these fibrils, paving the way for a comprehensive, non-invasive biomarker platform for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Luisa Stöckli
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Ansgar Felbecker
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| |
Collapse
|
2
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
3
|
Dato C, Micaglio E, Moresco G, Rondinone O, Vitali P, Pappone C, Fontana L, Miozzo M, Bet L. Case report: Chorea and cognitive decline in a young woman: instrumental and genetic assessment of a case originally diagnosed as multiple sclerosis. Front Genet 2023; 14:1129289. [PMID: 37434948 PMCID: PMC10331612 DOI: 10.3389/fgene.2023.1129289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/04/2023] [Indexed: 07/13/2023] Open
Abstract
We describe the case of a young woman affected by debilitating chorea and rapidly progressive cognitive decline. While her original diagnosis was multiple sclerosis, we performed a full instrumental and genetic assessement, though which we identified multiple genetic variants, including a novel variant of the APP gene. We propose some possible mechanisms by which such variants may contribute to neuroinflammation and ultimately lead to this devastating clinical course.
Collapse
Affiliation(s)
- Clemente Dato
- Unit of Neurology and Stroke Unit, IRCCS Policlinico San Donato, Milan, Italy
- Department of Neurology, Azienda Ospedaliera di Melegnano e Della Martesana, Melegnano, Italy
| | - Emanuele Micaglio
- Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Giada Moresco
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Ornella Rondinone
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Paolo Vitali
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Unit of Radiology, IRCCS Policlinico, San Donato, Milan, Italy
| | - Carlo Pappone
- Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Laura Fontana
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Monica Miozzo
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Luciano Bet
- Unit of Neurology and Stroke Unit, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
4
|
Patra P, Rani A, Sharma N, Mukherjee C, Jha HC. Unraveling the Connection of Epstein-Barr Virus and Its Glycoprotein M 146-157 Peptide with Neurological Ailments. ACS Chem Neurosci 2023. [PMID: 37290090 DOI: 10.1021/acschemneuro.3c00231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Epstein-Barr virus (EBV) is known to be associated with several cancers along with neurological modalities like Alzheimer's disease (AD) and multiple sclerosis (MS). Previous study from our group revealed that a 12 amino acid peptide fragment (146SYKHVFLSAFVY157) of EBV glycoprotein M (gM) exhibits amyloid-like self-aggregative properties. In the current study, we have investigated its effect on Aβ42 aggregation along with its effect on neural cell immunology and disease markers. EBV virion was also considered for the above-mentioned investigation. An increase in the aggregation of Aβ42 peptide was observed upon incubation with gM146-157. Further, the exposure of EBV and gM146-157 onto neuronal cells indicated the upregulation of inflammatory molecules like IL-1β, IL-6, TNF-α, and TGF-β that suggested neuroinflammation. Besides, host cell factors like mitochondrial potential and calcium ion signaling play a crucial role in cellular homeostasis and alterations in these factors aid in neurodegeneration. Changes in mitochondrial membrane potential manifested a decrease while elevation in the level of total Ca2+ ions was observed. Amelioration of Ca2+ ions triggers excitotoxicity in neurons. Subsequently, neurological disease-associated genes APP, ApoE4, and MBP were found to be increased at the protein level. Additionally, demyelination of neurons is a hallmark of MS and the myelin sheath consists of ∼70% of lipid/cholesterol-associated moieties. Hereby, genes associated with cholesterol metabolism indicated changes at the mRNA level. Enhanced expression of neurotropic factors like NGF and BDNF was discerned postexposure to EBV and gM146-157. Altogether, this study delineates a direct connection of EBV and its peptide gM146-157 with neurological illnesses.
Collapse
Affiliation(s)
- Priyanka Patra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| | - Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| | - Neha Sharma
- Department of Atomic Energy, Optical Coatings Laboratory, High Energy Lasers & Optics Section, Laser Technology Division, Laser Group, Raja Ramanna Centre for Advanced Technology, Indore 452013, Madhya Pradesh, India
| | - Chandrachur Mukherjee
- Department of Atomic Energy, Optical Coatings Laboratory, High Energy Lasers & Optics Section, Laser Technology Division, Laser Group, Raja Ramanna Centre for Advanced Technology, Indore 452013, Madhya Pradesh, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| |
Collapse
|
5
|
Petitfour J, Ayrignac X, Ginestet N, Prin P, Carra-Dallière C, Hirtz C, Charif M, Lehmann S, Labauge P. CSF β-amyloid is not a prognostic marker in multiple sclerosis patients. Mult Scler Relat Disord 2022; 68:104096. [PMID: 36037751 DOI: 10.1016/j.msard.2022.104096] [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: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating disorder. Given its variable prognosis, the identification of new prognostic biomarkers is needed. OBJECTIVES The aims of our study were to assess the prognostic values of CSF β-amyloid-42 (Aβ42) and β-amyloid-40 (Aβ40) levels in MS patients. METHODS Eighty-nine (55 RRMS, 34 PPMS) patients with a recent diagnosis and 27 controls were included in this single-centre retrospective study. Clinical, MRI and CSF data have been collected and were analysed to evaluate the potential value of CSF Aβ42 and Aβ40 levels as MS biomarkers. RESULTS CSF Aβ levels as well as Aβ42/Aβ40 ratio were identical in MS patients and controls. Although CSF Aβ42 and Aβ40 levels were higher in PPMS than in RRMS and in patients with higher EDSS, a multivariate analysis including age and EDSS demonstrated that only age of patients was associated with CSF amyloid levels. Additionally, 55 RRMS patients were followed for 3 years. We found no association between baseline amyloid levels and 3-year disability. CONCLUSION Our data do not support an association between CSF amyloid levels and MS status and disease severity. We suggest that CSF amyloid levels are not a prognostic biomarker in recently diagnosed RRMS.
Collapse
Affiliation(s)
- Justine Petitfour
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France
| | - Xavier Ayrignac
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France.
| | - Nelly Ginestet
- Univ Montpellier, INM, IRMB, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Pauline Prin
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France
| | - Clarisse Carra-Dallière
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France
| | - Christophe Hirtz
- Univ Montpellier, INM, IRMB, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Mahmoud Charif
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France
| | - Sylvain Lehmann
- Univ Montpellier, INM, IRMB, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Pierre Labauge
- Département de Neurologie, Univ Montpellier, INM, INSERM, MS Referral Centre & Reference Centre for Adult-Onset Leukodystrophies, CHU Montpellier, 80 Av Augustin Fliche, Montpellier 34295, France
| |
Collapse
|
6
|
Padovani A, Canale A, Schiavon L, Masciocchi S, Imarisio A, Risi B, Bonzi G, De Giuli V, Di Luca M, Ashton NJ, Blennow K, Zetterberg H, Pilotto A. Is amyloid involved in acute neuroinflammation? A CSF analysis in encephalitis. Alzheimers Dement 2022; 18:2167-2175. [PMID: 35084105 PMCID: PMC9787884 DOI: 10.1002/alz.12554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/07/2021] [Accepted: 10/25/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Several investigations have argued for a strong relationship between neuroinflammation and amyloid metabolism but it is still unclear whether inflammation exerts a pro-amyloidogenic effect, amplifies the neurotoxic effect of amyloid, or is protective. METHODS Forty-two patients with acute encephalitis (ENC) and 18 controls underwent an extended cerebrospinal fluid (CSF) panel of inflammatory, amyloid (Aβ40, 42, and 38, sAPP-α, sAPP-β), glial, and neuronal biomarkers. Linear and non-linear correlations between CSF biomarkers were evaluated studying conditional independence relationships. RESULTS CSF levels of inflammatory cytokines and neuronal/glial markers were higher in ENC compared to controls, whereas the levels of amyloid-related markers did not differ. Inflammatory markers were not associated with amyloid markers but exhibited a correlation with glial and neuronal markers in conditional independence analysis. DISCUSSION By an extensive CSF biomarkers analysis, this study showed that an acute neuroinflammation state, which is associated with glial activation and neuronal damage, does not influence amyloid homeostasis.
Collapse
Affiliation(s)
- Alessandro Padovani
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Antonio Canale
- Department of Statistical SciencesUniversity of PadovaPadovaItaly
| | - Lorenzo Schiavon
- Department of Statistical SciencesUniversity of PadovaPadovaItaly
| | - Stefano Masciocchi
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Alberto Imarisio
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Barbara Risi
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Giulio Bonzi
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | | | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden,Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden,Institute of PsychiatryPsychology and NeuroscienceMaurice Wohl Institute Clinical Neuroscience InstituteKing's College LondonLondonUK,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience & PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden,Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden,Department of Neurodegenerative DiseasesUCL Institute of NeurologyLondonUK,UK Dementia Research Institute at UCLLondonUK
| | - Andrea Pilotto
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| |
Collapse
|
7
|
Zetterberg H. Biofluid-based biomarkers for Alzheimer's disease-related pathologies: An update and synthesis of the literature. Alzheimers Dement 2022; 18:1687-1693. [PMID: 35213777 PMCID: PMC9514308 DOI: 10.1002/alz.12618] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/05/2021] [Accepted: 01/10/2022] [Indexed: 01/24/2023]
Abstract
The past few years have seen an explosion in sensitive and specific assays for cerebrospinal fluid (CSF) and blood biomarkers for Alzheimer's disease (AD) and related disorders, as well as some novel assays based on pathological seed-induced protein misfolding in patient samples. Here, I review this exciting field that promises to transform dementia diagnostics and disease monitoring. I discuss data on biomarkers for amyloid beta (Aβ) and tau pathology, neurodegeneration, and glial activation, mention the most promising biomarkers for α-synuclein and TDP-43 pathology, and highlight the need for further research into common co-pathologies. Finally, I consider practical aspects of blood-based biomarker-supported AD diagnostics and emphasize the importance of biomarker interpretation in a full clinical context.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| |
Collapse
|
8
|
Kalle J, Pontus W, Lenka N, Simon S, Ann B, Gunnar B, Kaj B, Henrik Z, Markus A. Cerebrospinal fluid amyloid precursor protein as a potential biomarker of fatigue in multiple sclerosis: A pilot study. Mult Scler Relat Disord 2022; 63:103846. [DOI: 10.1016/j.msard.2022.103846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/15/2022] [Accepted: 05/01/2022] [Indexed: 11/28/2022]
|
9
|
Nerattini M, Rubino F, Arnone A, Polito C, Mazzeo S, Lombardi G, Puccini G, Nacmias B, De Cristofaro MT, Sorbi S, Pupi A, Sciagrà R, Bessi V, Berti V. Cerebral amyloid load determination in a clinical setting: interpretation of amyloid biomarker discordances aided by tau and neurodegeneration measurements. Neurol Sci 2021; 43:2469-2480. [PMID: 34739618 DOI: 10.1007/s10072-021-05704-2] [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: 06/21/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) diagnosis can be hindered by amyloid biomarkers discordances. OBJECTIVE We aim to interpret discordances between amyloid positron emission tomography (Amy-PET) and cerebrospinal fluid (CSF) (Aβ42 and Aβ42/40), using Amy-PET semiquantitative analysis, [18F]fluorodeoxyglucose (FDG)-PET pattern, and CSF assays. METHOD Thirty-six subjects with dementia or mild cognitive impairment, assessed by neuropsychological tests, structural and functional imaging, and CSF assays (Aβ42, Aβ42/40, p-tau, t-tau), were retrospectively examined. Amy-PET and FDG-PET scans were analyzed by visual assessment and voxel-based analysis. SUVR were calculated on Amy-PET scans. RESULTS Groups were defined basing on the agreement among CSF Aβ42 (A), CSF Aβ42/40 Ratio (R), and Amy-PET (P) dichotomic results ( ±). In discordant groups, CSF assays, Amy-PET semiquantification, and FDG-PET patterns supported the diagnosis suggested by any two agreeing amyloid biomarkers. In groups with discordant CSF Aβ42, the ratio always agrees with Amy-PET results, solving both false-negative and false-positive Aβ42 results, with Aβ42 levels close to the cut-off in A + R-P- subjects. The A + R + P- group presented high amyloid deposition in relevant areas, such as precuneus, posterior cingulate cortex (PCC) and dorsolateral frontal inferior cortex at semiquantitative analysis. CONCLUSION The amyloid discordant cases could be overcome by combining CSF Aβ42, CSF ratio, and Amy-PET results. The concordance of any 2 out of the 3 biomarkers seems to reveal the remaining one as a false result. A cut-off point review could avoid CSF Aβ42 false-negative results. The regional semiquantitative Amy-PET analysis in AD areas, such as precuneus and PCC, could increase the accuracy in AD diagnosis.
Collapse
Affiliation(s)
- Matilde Nerattini
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy.
| | - Federica Rubino
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Annachiara Arnone
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Cristina Polito
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Gemma Lombardi
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Giulia Puccini
- Department of Nuclear Medicine, Hospital of Prato, Via Suor Niccolina Infermiera, 20/22, 59100, Prato, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Maria Teresa De Cristofaro
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143, Florence, Italy
| | - Alberto Pupi
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Roberto Sciagrà
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Valentina Berti
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139, Florence, Italy
| |
Collapse
|
10
|
Baiardi S, Pizza F, Polischi B, Moresco M, Abu-Rumeileh S, Plazzi G, Parchi P. Cerebrospinal fluid biomarkers of neurodegeneration in narcolepsy type 1. Sleep 2021; 43:5573415. [PMID: 31552425 DOI: 10.1093/sleep/zsz215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/27/2019] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES To measure the levels of five neurodegenerative biomarkers in the cerebrospinal fluid (CSF) of patients with narcolepsy type 1 (NT1) with variable disease duration. METHODS Following a standardized protocol of CSF collection and storage, we measured CSF total- and phosphorylated-tau, amyloid-beta 1-40 and 1-42, and neurofilament light chain (NfL) proteins in 30 nonneurological controls and 36 subjects with NT1, including 14 patients with recent disease onset (i.e. ≤12 months, short disease duration group). RESULTS CSF levels of all biomarkers were similar in NT1 subjects and controls. The comparison between NT1 with short and long disease duration only revealed slightly higher levels of CSF amyloid-beta 1-40 in the former group (median 9,549.5, interquartile range [IQR] 7,064.2-11,525.0 vs. 6,870.0, IQR 5,133.7-9,951.2, p = 0.043). CSF storage time did not influence the levels of the tested biomarkers. CONCLUSIONS The measurement of CSF total-tau, phosphorylated-tau, amyloid-beta 1-40 and 1-42, and NfL proteins is not informative in NT1.
Collapse
Affiliation(s)
- Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Fabio Pizza
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Barbara Polischi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Monica Moresco
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Samir Abu-Rumeileh
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Plazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Gaetani L, Salvadori N, Chipi E, Gentili L, Borrelli A, Parnetti L, Di Filippo M. Cognitive impairment in multiple sclerosis: lessons from cerebrospinal fluid biomarkers. Neural Regen Res 2021; 16:36-42. [PMID: 32788445 PMCID: PMC7818856 DOI: 10.4103/1673-5374.286949] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment is a common clinical manifestation of multiple sclerosis, but its pathophysiology is not completely understood. White and grey matter injury together with synaptic dysfunction do play a role. The measurement of biomarkers in the cerebrospinal fluid and the study of their association with cognitive impairment may provide interesting in vivo evidence of the biological mechanisms underlying multiple sclerosis-related cognitive impairment. So far, only a few studies on this topic have been published, giving interesting results that deserve further investigation. Cerebrospinal fluid biomarkers of different pathophysiological mechanisms seem to reflect different neuropsychological patterns of cognitive deficits in multiple sclerosis. The aim of this review is to discuss the studies that have correlated cerebrospinal fluid markers of immune, glial and neuronal pathology with cognitive impairment in multiple sclerosis. Although preliminary, these findings suggest that cerebrospinal fluid biomarkers show some correlation with cognitive performance in multiple sclerosis, thus providing interesting insights into the mechanisms underlying the involvement of specific cognitive domains.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucia Gentili
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Angela Borrelli
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
12
|
Porter L, Shoushtarizadeh A, Jelinek GA, Brown CR, Lim CK, de Livera AM, Jacobs KR, Weiland TJ. Metabolomic Biomarkers of Multiple Sclerosis: A Systematic Review. Front Mol Biosci 2020; 7:574133. [PMID: 33381517 PMCID: PMC7768024 DOI: 10.3389/fmolb.2020.574133] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Magnetic resonance imaging (MRI), cerebrospinal fluid (CSF) analysis, and the McDonald’s clinical criteria are currently utilized tools in diagnosing multiple sclerosis. However, a more conclusive, consistent, and efficient way of diagnosing multiple sclerosis (MS) is yet to be discovered. A potential biomarker, discovered using advances in high-throughput sequencing such as nuclear magnetic resonance (NMR) spectroscopy and other “Omics”-based techniques, may make diagnosis and prognosis more reliable resulting in a more personalized and targeted treatment regime and improved outcomes. The aim of this review was to systematically search the literature for potential biomarkers from any bodily fluid that could consistently and accurately diagnose MS and/or indicate disease progression. Methods A systematic literature review of EMBASE, PubMed (MEDLINE), The Cochrane Library, and CINAHL databases produced over a thousand potential studies. Inclusion criteria stated studies with potential biomarker outcomes for people with MS were to be included in the review. Studies were limited to those with human participants who had a clinically defined diagnosis of MS and published in English, with no limit placed on date of publication or the type of bodily fluid sampled. Results A total of 1,805 studies were recorded from the literature search. A total of 1,760 studies were removed based on their abstract, with a further 18 removed after considering the full text. A total of 30 studies were considered relevant and had their data retrieved and analyzed. Due to the heterogeneity of focus and results from the refined studies, a narrative synthesis was favored. Conclusion Several promising candidate biomarkers suitable for clinical application in MS have been studied. It is recommended follow-up studies with larger sample sizes be completed on several potential biomarkers.
Collapse
Affiliation(s)
- Lachlan Porter
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Alireza Shoushtarizadeh
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - George A Jelinek
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Chelsea R Brown
- The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Chai K Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Alysha M de Livera
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Kelly R Jacobs
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Tracey J Weiland
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
13
|
Stojić-Vukanić Z, Hadžibegović S, Nicole O, Nacka-Aleksić M, Leštarević S, Leposavić G. CD8+ T Cell-Mediated Mechanisms Contribute to the Progression of Neurocognitive Impairment in Both Multiple Sclerosis and Alzheimer's Disease? Front Immunol 2020; 11:566225. [PMID: 33329528 PMCID: PMC7710704 DOI: 10.3389/fimmu.2020.566225] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Neurocognitive impairment (NCI) is one of the most relevant clinical manifestations of multiple sclerosis (MS). The profile of NCI and the structural and functional changes in the brain structures relevant for cognition in MS share some similarities to those in Alzheimer's disease (AD), the most common cause of neurocognitive disorders. Additionally, despite clear etiopathological differences between MS and AD, an accumulation of effector/memory CD8+ T cells and CD8+ tissue-resident memory T (Trm) cells in cognitively relevant brain structures of MS/AD patients, and higher frequency of effector/memory CD8+ T cells re-expressing CD45RA (TEMRA) with high capacity to secrete cytotoxic molecules and proinflammatory cytokines in their blood, were found. Thus, an active pathogenetic role of CD8+ T cells in the progression of MS and AD may be assumed. In this mini-review, findings supporting the putative role of CD8+ T cells in the pathogenesis of MS and AD are displayed, and putative mechanisms underlying their pathogenetic action are discussed. A special effort was made to identify the gaps in the current knowledge about the role of CD8+ T cells in the development of NCI to "catalyze" translational research leading to new feasible therapeutic interventions.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Senka Hadžibegović
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Olivier Nicole
- Institut des Maladies Neurodégénératives, CNRS, UMR5293, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR5293, Bordeaux, France
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Sanja Leštarević
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
14
|
Medical Histories of Control Subjects Influence the Biomarker Potential of Plasma Aβ in Alzheimer's Disease: a Meta-analysis. J Mol Neurosci 2020; 70:861-870. [PMID: 32125624 DOI: 10.1007/s12031-020-01510-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Whether blood amyloid-β (Aβ) could be a peripheral biomarker of Alzheimer's disease (AD) remains in dispute. In the present study, we conducted a meta-analysis with 19 citations searched from Embase, PubMed, and the Cochrane Library database. Weighted mean difference (WMD) with 95% confidence intervals (CIs) was used to estimate the effect size. We firstly analyzed the plasma Aβ40, Aβ42, and Aβ42/Aβ40 ratio in AD and control group subjects. However, only a lower level of plasma Aβ42 was figured out in AD group subjects with weak statistical significance (WMD 1.82; 95% CI 0.59, 3.06; P = 0.004; I2 = 84%). We considered that the medical histories of control subjects could influence the biomarker ability of plasma Aβ. Therefore, subgroup analyses were then carried out based on a new recruiting criterion for control subjects, defining as no afflictions of any Aβ-related diseases. Surprisingly, AD group subjects showed a significant decrease in plasma Aβ42/Aβ40 ratio with low heterogeneity among studies (WMD 0.02; 95% CI 0.02, 0.02; P < 0.00001; I2 = 0%). Moreover, not only the Aβ42/Aβ40 ratio but also Aβ42 and Aβ40 were indifferent between AD and pseudo-control subjects which might be afflicted with Aβ-related diseases. This meta-analysis demonstrated that medical histories of control subjects were interference factors impeding plasma Aβ to be a biomarker of AD.
Collapse
|
15
|
CSF β-amyloid predicts early cerebellar atrophy and is associated with a poor prognosis in multiple sclerosis. Mult Scler Relat Disord 2020; 37:101462. [DOI: 10.1016/j.msard.2019.101462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/17/2019] [Accepted: 10/20/2019] [Indexed: 12/29/2022]
|
16
|
Pietroboni AM, Colombi A, Carandini T, Contarino VE, Ghezzi L, Fumagalli GG, Arighi A, Fenoglio C, De Riz MA, Triulzi F, Scarpini E, Galimberti D. Low CSF β-amyloid levels predict early regional grey matter atrophy in multiple sclerosis. Mult Scler Relat Disord 2019; 39:101899. [PMID: 31884385 DOI: 10.1016/j.msard.2019.101899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE Grey matter (GM) atrophy is present from the earliest stages of multiple sclerosis (MS) and occurs largely in a nonrandom manner. However, the biological mechanisms underlying the progression of regional atrophy are still unclear. Aim of this study is to investigate whether amyloid pathology might be involved in determining the pattern of GM atrophy over time. METHODS Forty-six subjects were recruited: 31 newly diagnosed relapsing-remitting (RR-) MS patients and 15 age- and sex-matched healthy controls (HC). Aβ levels were determined in CSF samples from all subjects. All participants underwent brain magnetic resonance imaging (MRI) at baseline, and 23 out of 31 patients at one year follow-up. T1-weighted scans were segmented using the Geodesic Information Flows software. Non-parametric statistical tests were used for between-group comparisons and multiple regression analyses. RESULTS CSF Aβ concentration was the best predictor of global GM loss over time after age (β = 0.403; p = 0.024), in particular in the left precuneus (p = 0.045), in the left middle cingulate gyrus (p = 0.009), in the left precentral gyrus (p = 0.021) and in the right angular gyrus (p = 0.029). CONCLUSIONS CSF Aβ levels seem to be crucial in MS early brain volume loss as GM atrophy manifests in regions particularly vulnerable to early Aβ deposition.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy.
| | - Annalisa Colombi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | | | - Laura Ghezzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Giorgio G Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Andrea Arighi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Chiara Fenoglio
- Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Milena A De Riz
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Fabio Triulzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; University of Milan, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Dino Ferrari Center, Milan, Italy; University of Milan, Milan, Italy
| |
Collapse
|
17
|
Illán-Gala I, Pegueroles J, Montal V, Alcolea D, Vilaplana E, Bejanin A, Borrego-Écija S, Sampedro F, Subirana A, Sánchez-Saudinós MB, Rojas-García R, Vanderstichele H, Blesa R, Clarimón J, Antonell A, Lladó A, Sánchez-Valle R, Fortea J, Lleó A. APP-derived peptides reflect neurodegeneration in frontotemporal dementia. Ann Clin Transl Neurol 2019; 6:2518-2530. [PMID: 31789459 PMCID: PMC6917306 DOI: 10.1002/acn3.50948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Objective We aimed to investigate the relationship between cerebrospinal fluid levels (CSF) of amyloid precursor protein (APP)‐derived peptides related to the amyloidogenic pathway, cortical thickness, neuropsychological performance, and cortical gene expression profiles in frontotemporal lobar degeneration (FTLD)‐related syndromes, Alzheimer’s disease (AD), and healthy controls. Methods We included 214 participants with CSF available recruited at two centers: 93 with FTLD‐related syndromes, 57 patients with AD, and 64 healthy controls. CSF levels of amyloid β (Aβ)1‐42, Aβ1‐40, Aβ1‐38, and soluble β fragment of APP (sAPPβ) were centrally analyzed. We compared CSF levels of APP‐derived peptides between groups and, we studied the correlation between CSF biomarkers, cortical thickness, and domain‐specific cognitive composites in each group. Then, we explored the relationship between cortical thickness, CSF levels of APP‐derived peptides, and regional gene expression profile using a brain‐wide regional gene expression data in combination with gene set enrichment analysis. Results The CSF levels of Aβ1‐40, Aβ1‐38, and sAPPβ were lower in the FTLD‐related syndromes group than in the AD and healthy controls group. CSF levels of all APP‐derived peptides showed a positive correlation with cortical thickness and the executive cognitive composite in the FTLD‐related syndromes group but not in the healthy control or AD groups. In the cortical regions where we observed a significant association between cortical thickness and CSF levels of APP‐derived peptides, we found a reduced expression of genes related to synaptic function. Interpretation APP‐derived peptides in CSF may reflect FTLD‐related neurodegeneration. This observation has important implications as Aβ1‐42 levels are considered an indirect biomarker of cerebral amyloidosis.
Collapse
Affiliation(s)
- Ignacio Illán-Gala
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Jordi Pegueroles
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Eduard Vilaplana
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alexandre Bejanin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders, Neurology Department, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica, Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Andrea Subirana
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María-Belén Sánchez-Saudinós
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Rafael Blesa
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Jordi Clarimón
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders, Neurology Department, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica, Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders, Neurology Department, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica, Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders, Neurology Department, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica, Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Juan Fortea
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain.,Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| |
Collapse
|
18
|
Blennow K, Zetterberg H. Fluid biomarker-based molecular phenotyping of Alzheimer's disease patients in research and clinical settings. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:3-23. [PMID: 31699324 DOI: 10.1016/bs.pmbts.2019.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is very difficult to diagnose on pure clinical grounds, especially in the earlier phases of the disease. At the same time, lessons from recent clinical trials suggest that treatments have to be initiated very early, to have a chance to show clinical efficacy. Therefore, biomarkers reflecting core AD pathophysiology have a key position in clinical trials and clinical management. The core AD cerebrospinal fluid (CSF) biomarker toolbox include amyloid β (Aβ42 and the Aβ42/40 ratio) reflecting brain amyloidosis, total tau (T-tau) reflecting neurodegeneration intensity, and phosphorylated tau (P-tau) that is related to tau pathology. These CSF biomarkers have very consistently been found to have high diagnostic accuracy, also in earlier disease stages. Importantly, CSF Aβ42 and Aβ42/40 ratio show excellent agreement with amyloid PET readouts, indicating that these biomarker tests can be used interchangeably. Intense collaborative standardization efforts have given Certified Reference Materials (CRMs) to harmonize assay formats for CSF Aβ42, the most central AD biomarker, and CRMs for Aβ40 are under development. The core AD biomarkers are today available on high-precision fully automated analytical platforms, which will serve to introduce uniform cut-off levels and enable the large-scale introduction of CSF biomarkers for routine disease diagnosis. Of novel biomarker candidates, synaptic proteins, such as the dendritic protein neurogranin, show promise as tools to monitor synaptic degeneration, an important aspect of AD pathophysiology. Recent studies show that the core AD biomarkers also can be measured in blood samples. Ultra-sensitive assays that allow for quantification of neuronal proteins, such as tau and neurofilament light (NFL) in blood samples. Further, plasma Aβ42 and Aβ42/40 show high concordance with brain amyloidosis evaluated by PET scans. In the future, blood biomarkers may have value as screening tools, especially to rule out patients without biomarker evidence of AD pathology.
Collapse
Affiliation(s)
- Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology;the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology;the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, United Kingdom; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom.
| |
Collapse
|
19
|
Tackenberg C, Nitsch RM. The secreted APP ectodomain sAPPα, but not sAPPβ, protects neurons against Aβ oligomer-induced dendritic spine loss and increased tau phosphorylation. Mol Brain 2019; 12:27. [PMID: 30922360 PMCID: PMC6440141 DOI: 10.1186/s13041-019-0447-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
Aim The amyloid precursor protein (APP) is endoproteolytically processed to generate either the neurotoxic beta-amyloid peptide (Aβ) or the secreted ectodomain APP alpha (sAPPα). While neurotrophic properties of sAPPα were suggested in several studies, it is still unclear if and how sAPPα counteracts pathogenic effects of Aβ. Direct comparisons with sAPPβ, produced in the Aβ-generating pathway, are missing in order to determine the role of sAPPα’s carbonyl-terminal end in its possible neuroprotective activity. Methods Mouse neuronal primary cultures and hippocampal slices were treated with oligomeric Aβ42. The effects on tau phosphorylation and dendritic spine densities were assessed by western blot and confocal imaging, respectively. Co-administration of either sAPPα or sAPPβ was used to determine activity on Aβ-induced toxicity. Results/discussion We found that oligomeric Aβ strongly increased AT8 and AT180 phosphorylation of tau and caused a loss of dendritic spines. SAPPα completely abolished Aβ effects whereas sAPPβ had no such rescue activity. Interestingly, sAPPα or sAPPβ alone neither affected tau phosphorylation nor dendritic spine numbers. Together, our data suggest that sAPPα specifically protects neurons against Aβ-dependent toxicity supporting the strategy of activating α-secretase-dependent endoproteolytic APP processing to increase sAPPα shedding from the neuronal plasma membrane as a therapeutic intervention for the protection of dendritic spines and phospho-tau-dependent toxicity in Alzheimer’s disease. Electronic supplementary material The online version of this article (10.1186/s13041-019-0447-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland. .,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland.
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Kristofikova Z, Ricny J, Kaping D, Klaschka J, Kotoucova J, Bartos A. Levels of 17β-hydroxysteroid dehydrogenase type 10 in CSF are not a valuable biomarker for multiple sclerosis. Biomark Med 2018; 12:1331-1340. [DOI: 10.2217/bmm-2018-0061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We aimed to characterize the role of mitochondrial 17β-hydroxysteroid dehydrogenase type 10 (17β-HSD10) overexpression in multiple sclerosis (MS) and to evaluate its use as a biomarker. Materials & methods: We estimated levels of 17β-HSD10, amyloid β 1–42, cyclophilin D, 17β-HSD10-cyclophilin D complexes or 17β-HSD10-parkin complexes in cerebrospinal fluid (CSF) samples. Results: The increase in 17β-HSD10 levels or in 17β-HSD10-parkin complexes and links to leukocytes were found only in relapsing–remitting MS. The sensitivity of the biomarker was 64%, the specificity equaled 60–63% compared with controls. Conclusion: Increased CSF levels of 17β-HSD10 in later stages of MS could be interpreted via its upregulation in demyelinated neuronal axons. CSF levels of 17β-HSD10 are not the valuable biomarker for the early diagnosis or for the progression of MS.
Collapse
Affiliation(s)
| | - Jan Ricny
- National Institute of Mental Health, 250 67 Klecany, Czech Republic
| | - Daniel Kaping
- National Institute of Mental Health, 250 67 Klecany, Czech Republic
| | - Jan Klaschka
- Institute of Computer Science, Academy of Sciences, 182 07 Prague, Czech Republic
| | - Jolana Kotoucova
- National Institute of Mental Health, 250 67 Klecany, Czech Republic
| | - Ales Bartos
- National Institute of Mental Health, 250 67 Klecany, Czech Republic
- Department of Neurology, Third Faculty of Medicine, University Hospital Kralovske Vinohrady, Charles University, 100 34 Prague, Czech Republic
| |
Collapse
|
21
|
Pietroboni AM, Carandini T, Colombi A, Mercurio M, Ghezzi L, Giulietti G, Scarioni M, Arighi A, Fenoglio C, De Riz MA, Fumagalli GG, Basilico P, Serpente M, Bozzali M, Scarpini E, Galimberti D, Marotta G. Amyloid PET as a marker of normal-appearing white matter early damage in multiple sclerosis: correlation with CSF β-amyloid levels and brain volumes. Eur J Nucl Med Mol Imaging 2018; 46:280-287. [PMID: 30343433 DOI: 10.1007/s00259-018-4182-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE The disease course of multiple sclerosis (MS) is unpredictable, and reliable prognostic biomarkers are needed. Positron emission tomography (PET) with β-amyloid tracers is a promising tool for evaluating white matter (WM) damage and repair. Our aim was to investigate amyloid uptake in damaged (DWM) and normal-appearing WM (NAWM) of MS patients, and to evaluate possible correlations between cerebrospinal fluid (CSF) β-amyloid1-42 (Aβ) levels, amyloid tracer uptake, and brain volumes. METHODS Twelve MS patients were recruited and divided according to their disease activity into active and non-active groups. All participants underwent neurological examination, neuropsychological testing, lumbar puncture, brain magnetic resonance (MRI) imaging, and 18F-florbetapir PET. Aβ levels were determined in CSF samples from all patients. MRI and PET images were co-registered, and mean standardized uptake values (SUV) were calculated for each patient in the NAWM and in the DWM. To calculate brain volumes, brain segmentation was performed using statistical parametric mapping software. Nonparametric statistical analyses for between-group comparisons and regression analyses were conducted. RESULTS We found a lower SUV in DWM compared to NAWM (p < 0.001) in all patients. Decreased NAWM-SUV was observed in the active compared to non-active group (p < 0.05). Considering only active patients, NAWM volume correlated with NAWM-SUV (p = 0.01). Interestingly, CSF Aβ concentration was a predictor of both NAWM-SUV (r = 0.79; p = 0.01) and NAWM volume (r = 0.81, p = 0.01). CONCLUSIONS The correlation between CSF Aβ levels and NAWM-SUV suggests that the predictive role of β-amyloid may be linked to early myelin damage and may reflect disease activity and clinical progression.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy. .,University of Milan, Milan, Italy. .,Dino Ferrari Center, Milan, Italy.
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Annalisa Colombi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Matteo Mercurio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Laura Ghezzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | | | - Marta Scarioni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Andrea Arighi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | | | - Milena A De Riz
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Giorgio G Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Paola Basilico
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | | | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy.,Dino Ferrari Center, Milan, Italy
| | - Giorgio Marotta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.,University of Milan, Milan, Italy
| |
Collapse
|
22
|
Pietroboni AM, Caprioli M, Carandini T, Scarioni M, Ghezzi L, Arighi A, Cioffi S, Cinnante C, Fenoglio C, Oldoni E, De Riz MA, Basilico P, Fumagalli GG, Colombi A, Giulietti G, Serra L, Triulzi F, Bozzali M, Scarpini E, Galimberti D. CSF β-amyloid predicts prognosis in patients with multiple sclerosis. Mult Scler 2018; 25:1223-1231. [PMID: 30084711 DOI: 10.1177/1352458518791709] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The importance of predicting disease progression in multiple sclerosis (MS) has increasingly been recognized, and hence reliable biomarkers are needed. OBJECTIVES To investigate the prognostic role of cerebrospinal fluid (CSF) amyloid beta1-42 (Aβ) levels by the determination of a cut-off value to classify patients in slow and fast progressors. To evaluate possible association with white matter (WM) and grey matter (GM) damage at early disease stages. METHODS Sixty patients were recruited and followed up for 3-5 years. Patients underwent clinical assessment, brain magnetic resonance imaging (MRI; at baseline and after 1 year), and CSF analysis to determine Aβ levels. T1-weighted volumes were calculated. T2-weighted scans were used to quantify WM lesion loads. RESULTS Lower CSF Aβ levels were observed in patients with a worse follow-up Expanded Disability Status Scale (EDSS; r = -0.65, p < 0.001). The multiple regression analysis confirmed CSF Aβ concentration as a predictor of patients' EDSS increase (r = -0.59, p < 0.0001). Generating a receiver operating characteristic curve, a cut-off value of 813 pg/mL was determined as the threshold able to identify patients with worse prognosis (95% confidence interval (CI): 0.690-0.933, p = 0.0001). No differences in CSF tau and neurofilament light chain (NfL) levels were observed (p > 0.05). CONCLUSION Low CSF Aβ levels may represent a predictive biomarker of disease progression in MS.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Michela Caprioli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Marta Scarioni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Laura Ghezzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Andrea Arighi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Sara Cioffi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Claudia Cinnante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | | | - Emanuela Oldoni
- Laboratory for Neuroimmunology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Milena A De Riz
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Paola Basilico
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Giorgio G Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy/Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy/University of Milan, Milan, Italy
| | - Annalisa Colombi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | | | - Laura Serra
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Fabio Triulzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy/ Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy/University of Milan, Dino Ferrari Center, Milan, Italy
| |
Collapse
|
23
|
|
24
|
Rostgaard N, Roos P, Portelius E, Blennow K, Zetterberg H, Simonsen AH, Nielsen JE. CSF neurofilament light concentration is increased in presymptomatic CHMP2B mutation carriers. Neurology 2017; 90:e157-e163. [PMID: 29237796 PMCID: PMC5772154 DOI: 10.1212/wnl.0000000000004799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022] Open
Abstract
Objective A rare cause of familial frontotemporal dementia (FTD) is a mutation in the CHMP2B gene on chromosome 3 (FTD-3), described in a Danish family. Here we examine whether CSF biomarkers change in the preclinical phase of the disease. Methods In this cross-sectional explorative study, we analyzed CSF samples from 16 mutation carriers and 14 noncarriers from the Danish FTD-3 family. CSF biomarkers included total tau (t-tau) and neurofilament light chain (NfL) as a marker for neurodegeneration, phosphorylated tau (p-tau) as a marker for tau pathology, β-amyloid (Aβ) 38, 40, and 42 (Aβ38, Aβ40, and Aβ42) to monitor Aβ metabolism, and YKL-40 as a marker of neuroinflammation. Aβ isoform concentrations were measured using a multiplexed immunoassay; t-tau, p-tau, NfL, and YKL-40 concentrations were measured using sandwich ELISAs. Results CSF NfL concentration was significantly increased in mutation carriers vs noncarriers. Further, CSF NfL concentration was significantly higher in symptomatic mutation carriers compared to presymptomatic carriers, and also significantly higher in presymptomatic carriers compared to noncarriers. No differences in t-tau and p-tau and YKL-40 concentrations between controls and mutation carriers were observed. CSF concentrations of the Aβ peptides Aβ38 and Aβ40 but not Aβ42 were significantly lower in mutation carriers compared to noncarriers. Conclusions Increased NfL levels in presymptomatic individuals and in symptomatic patients with FTD-3 indicate a continuous process of neurodegeneration from the presymptomatic to symptomatic state. Although not specific for FTD-3 pathology, our data suggest that CSF NfL could serve as a valuable biomarker to detect onset of neurodegeneration in FTD-3 mutation carriers.
Collapse
Affiliation(s)
- Nina Rostgaard
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Peter Roos
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Erik Portelius
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Henrik Zetterberg
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Anja H Simonsen
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| | - Jørgen E Nielsen
- From the Danish Dementia Research Centre (N.R., P.R., A.H.S., J.E.N.), Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark; Clinical Neurochemistry Laboratory (E.P., K.B., H.Z.), Sahlgrenska University Hospital; Institute of Neuroscience and Physiology (E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; and Department of Molecular Neuroscience and UK Dementia Research Institute (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
25
|
Stampanoni Bassi M, Garofalo S, Marfia GA, Gilio L, Simonelli I, Finardi A, Furlan R, Sancesario GM, Di Giandomenico J, Storto M, Mori F, Centonze D, Iezzi E. Amyloid-β Homeostasis Bridges Inflammation, Synaptic Plasticity Deficits and Cognitive Dysfunction in Multiple Sclerosis. Front Mol Neurosci 2017; 10:390. [PMID: 29209169 PMCID: PMC5702294 DOI: 10.3389/fnmol.2017.00390] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/08/2017] [Indexed: 11/13/2022] Open
Abstract
Cognitive deficits are frequently observed in multiple sclerosis (MS), mainly involving processing speed and episodic memory. Both demyelination and gray matter atrophy can contribute to cognitive deficits in MS. In recent years, neuroinflammation is emerging as a new factor influencing clinical course in MS. Inflammatory cytokines induce synaptic dysfunction in MS. Synaptic plasticity occurring within hippocampal structures is considered as one of the basic physiological mechanisms of learning and memory. In experimental models of MS, hippocampal plasticity is profoundly altered by proinflammatory cytokines. Although mechanisms of inflammation-induced hippocampal pathology in MS are not completely understood, alteration of Amyloid-β (Aβ) metabolism is emerging as a key factor linking together inflammation, synaptic plasticity and neurodegeneration in different neurological diseases. We explored the correlation between concentrations of Aβ1–42 and the levels of some proinflammatory and anti-inflammatory cytokines (interleukin-1β (IL-1β), IL1-ra, IL-8, IL-10, IL-12, tumor necrosis factor α (TNFα), interferon γ (IFNγ)) in the cerebrospinal fluid (CSF) of 103 remitting MS patients. CSF levels of Aβ1–42 were negatively correlated with the proinflammatory cytokine IL-8 and positively correlated with the anti-inflammatory molecules IL-10 and interleukin-1 receptor antagonist (IL-1ra). Other correlations, although noticeable, were either borderline or not significant. Our data show that an imbalance between proinflammatory and anti-inflammatory cytokines may lead to altered Aβ homeostasis, representing a key factor linking together inflammation, synaptic plasticity and cognitive dysfunction in MS. This could be relevant to identify novel therapeutic approaches to hinder the progression of cognitive dysfunction in MS.
Collapse
Affiliation(s)
- Mario Stampanoni Bassi
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Garofalo
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy
| | - Girolama A Marfia
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luana Gilio
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ilaria Simonelli
- Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Service of Medical Statistics & Information Technology, Fondazione Fatebenefratelli per la Ricerca e la Formazione Sanitaria e Sociale, Rome, Italy
| | - Annamaria Finardi
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Giulia M Sancesario
- Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Jonny Di Giandomenico
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy
| | - Marianna Storto
- Clinical Pathology Unit, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy
| | - Francesco Mori
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Diego Centonze
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology & Unit of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Pozzilli, Italy
| |
Collapse
|
26
|
Constantinides VC, Paraskevas GP, Emmanouilidou E, Petropoulou O, Bougea A, Vekrellis K, Evdokimidis I, Stamboulis E, Kapaki E. CSF biomarkers β-amyloid, tau proteins and a-synuclein in the differential diagnosis of Parkinson-plus syndromes. J Neurol Sci 2017; 382:91-95. [DOI: 10.1016/j.jns.2017.09.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/01/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022]
|
27
|
Earliest accumulation of β-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat Commun 2017; 8:1214. [PMID: 29089479 PMCID: PMC5663717 DOI: 10.1038/s41467-017-01150-x] [Citation(s) in RCA: 602] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/23/2017] [Indexed: 01/22/2023] Open
Abstract
It is not known exactly where amyloid-β (Aβ) fibrils begin to accumulate in individuals with Alzheimer’s disease (AD). Recently, we showed that abnormal levels of Aβ42 in cerebrospinal fluid (CSF) can be detected before abnormal amyloid can be detected using PET in individuals with preclinical AD. Using these approaches, here we identify the earliest preclinical AD stage in subjects from the ADNI and BioFINDER cohorts. We show that Aβ accumulation preferentially starts in the precuneus, medial orbitofrontal, and posterior cingulate cortices, i.e., several of the core regions of the default mode network (DMN). This early pattern of Aβ accumulation is already evident in individuals with normal Aβ42 in the CSF and normal amyloid PET who subsequently convert to having abnormal CSF Aβ42. The earliest Aβ accumulation is further associated with hypoconnectivity within the DMN and between the DMN and the frontoparietal network, but not with brain atrophy or glucose hypometabolism. Our results suggest that Aβ fibrils start to accumulate predominantly within certain parts of the DMN in preclinical AD and already then affect brain connectivity. Abnormal levels of Aβ42 in the cerebrospinal fluid occur prior to a positive amyloid PET scan in the brain of individuals with Alzheimer’s disease and here the authors use this temporal pattern to identify individuals with very early stage AD. They show that Aβ fibrils start to accumulate in some of the regions of the default mode network and affect brain connectivity before neurodegeneration occurs.
Collapse
|
28
|
Etemadifar M, Ghadimi M, Ghadimi K, Alsahebfosoul F. The Serum Amyloid β Level in Multiple Sclerosis: A Case- Control Study. CASPIAN JOURNAL OF NEUROLOGICAL SCIENCES 2017. [DOI: 10.29252/nirp.cjns.3.11.214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
29
|
Shahim P, Tegner Y, Marklund N, Höglund K, Portelius E, Brody DL, Blennow K, Zetterberg H. Astroglial activation and altered amyloid metabolism in human repetitive concussion. Neurology 2017; 88:1400-1407. [PMID: 28283595 DOI: 10.1212/wnl.0000000000003816] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/22/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether postconcussion syndrome (PCS) due to repetitive concussive traumatic brain injury (rcTBI) is associated with CSF biomarker evidence of astroglial activation, amyloid deposition, and blood-brain barrier (BBB) impairment. METHODS A total of 47 participants (28 professional athletes with PCS and 19 controls) were assessed with lumbar puncture (median 1.5 years, range 0.25-12 years after last concussion), standard MRI of the brain, and Rivermead Post-Concussion Symptoms Questionnaire (RPQ). The main outcome measures were CSF concentrations of astroglial activation markers (glial fibrillary acidic protein [GFAP] and YKL-40), markers reflecting amyloid precursor protein metabolism (Aβ38, Aβ40, Aβ42, sAPPα, and sAPPβ), and BBB function (CSF:serum albumin ratio). RESULTS Nine of the 28 athletes returned to play within a year, while 19 had persistent PCS >1 year. Athletes with PCS >1 year had higher RPQ scores and number of concussions than athletes with PCS <1 year. Median concentrations of GFAP and YKL-40 were higher in athletes with PCS >1 year compared with controls, although with an overlap between the groups. YKL-40 correlated with RPQ score and the lifetime number of concussions. Athletes with rcTBI had lower concentrations of Aβ40 and Aβ42 than controls. The CSF:serum albumin ratio was unaltered. CONCLUSIONS This study suggests that PCS may be associated with biomarker evidence of astroglial activation and β-amyloid (Aβ) dysmetabolism in the brain. There was no clear evidence of Aβ deposition as Aβ40 and Aβ42 were reduced in parallel. The CSF:serum albumin ratio was unaltered, suggesting that the BBB is largely intact in PCS.
Collapse
Affiliation(s)
- Pashtun Shahim
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| | - Yelverton Tegner
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Niklas Marklund
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kina Höglund
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Erik Portelius
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - David L Brody
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Henrik Zetterberg
- From the Institute of Neuroscience and Physiology (P.S., K.H., E.P., K.B., H.Z.), Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg; Clinical Neurochemistry Laboratory (P.S., K.H., E.P., K.B., H.Z.), Sahlgrenska University Hospital, Mölndal; Division of Medical Sciences, Department of Health Sciences (Y.T.), Luleå University of Technology; Department of Neuroscience, Neurosurgery (N.M.), Uppsala University, Uppsala, Sweden; Washington University School of Medicine (D.L.B.), St. Louis, MO; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
30
|
Singh J, Srivastava A, Sharma P, Pradhan P, Kundu B. DNA intercalators as amyloid assembly modulators: mechanistic insights. RSC Adv 2017. [DOI: 10.1039/c6ra26313e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
DNA intercalators modulate amyloid assembly of proteins through specific hetero-aromatic interactions diverting them to form amorphous aggregates.
Collapse
Affiliation(s)
- Jasdeep Singh
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Ankit Srivastava
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Pankaj Sharma
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Prashant Pradhan
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi
- India
| |
Collapse
|
31
|
Pietroboni AM, Schiano di Cola F, Scarioni M, Fenoglio C, Spanò B, Arighi A, Cioffi SM, Oldoni E, De Riz MA, Basilico P, Calvi A, Fumagalli GG, Triulzi F, Galimberti D, Bozzali M, Scarpini E. CSF β-amyloid as a putative biomarker of disease progression in multiple sclerosis. Mult Scler 2016; 23:1085-1091. [PMID: 27754941 DOI: 10.1177/1352458516674566] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Neurodegeneration plays a major role in determining disability in multiple sclerosis (MS) patients. Hence, there is increasing need to identify reliable biomarkers, which could serve as prognostic measure of disease progression. OBJECTIVES To assess whether cerebrospinal fluid (CSF) tau and β-amyloid (Aβ) levels were altered in newly diagnosed MS patients and correlated with disability. Moreover, we investigated whether these CSF biomarkers associate with macroscopic brain tissue damage measures. METHODS CSF Aβ and tau levels were determined by enzyme-linked immunosorbent assay in CSF samples from 48 newly diagnosed MS patients, followed-up clinically for 3 years by recording their Expanded Disability Status Scale score at 6-month intervals, and 45 controls. All patients underwent magnetic resonance imaging at baseline and at the end of follow-up to quantify their lesion load (LL). RESULTS CSF Aβ levels were significantly reduced in patients compared to controls ( p < 0.001). Lower CSF Aβ levels at baseline were a disability predictor at 3-year follow-up ( p = 0.009). CSF tau levels correlated with T2- and T1-LL ( p < 0.001). CONCLUSION CSF Aβ reduction is a promising biomarker of neurodegeneration and may predict patients' clinical outcome. Therefore, CSF Aβ should be considered as a potential biomarker of prognostic value.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Schiano di Cola
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Scarioni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Spanò
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Andrea Arighi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Mg Cioffi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Oldoni
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Milena A De Riz
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Basilico
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Calvi
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio G Fumagalli
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Triulzi
- Neuroradiology Unit, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Elio Scarpini
- Neurology Unit, Department of Pathophysiology and Transplantation, University of Milan, "Dino Ferrari" Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
32
|
Matías-Guiu JA, Oreja-Guevara C, Cabrera-Martín MN, Moreno-Ramos T, Carreras JL, Matías-Guiu J. Amyloid Proteins and Their Role in Multiple Sclerosis. Considerations in the Use of Amyloid-PET Imaging. Front Neurol 2016; 7:53. [PMID: 27065425 PMCID: PMC4814935 DOI: 10.3389/fneur.2016.00053] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 03/22/2016] [Indexed: 02/06/2023] Open
Abstract
Thioflavin T derivatives are used in positron-emission tomography (PET) studies to detect amyloid protein deposits in patients with Alzheimer disease. These tracers bind extensively to white matter, which suggests that they may be useful in studies of multiple sclerosis (MS), and that proteins resulting from proteolytic processing of the amyloid precursor protein (APP) may contribute to MS. This article reviews data from both clinical and preclinical studies addressing the role of these proteins, whether they are detected in CSF studies or using PET imaging. APP is widely expressed in demyelinated axons and may have a protective effect in MS and in experimental allergic encephalomyelitis in animals. Several mechanisms associated with this increased expression may affect the degree of remyelination in MS. Amyloid-PET imaging may help determine the degree of demyelination and provide information on the molecular changes linked to APP proteolytic processing experienced by patients with MS.
Collapse
Affiliation(s)
- Jordi A Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Teresa Moreno-Ramos
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| | - Jorge Matías-Guiu
- Department of Neurology, Hospital Clínico San Carlos, San Carlos Institute for Health Research (IdISSC), Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
33
|
Role of amyloid-β CSF levels in cognitive deficit in MS. Clin Chim Acta 2015; 449:23-30. [DOI: 10.1016/j.cca.2015.01.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/21/2015] [Indexed: 11/18/2022]
|
34
|
Neural Plasticity in Multiple Sclerosis: The Functional and Molecular Background. Neural Plast 2015; 2015:307175. [PMID: 26229689 PMCID: PMC4503575 DOI: 10.1155/2015/307175] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/09/2015] [Accepted: 06/21/2015] [Indexed: 01/19/2023] Open
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disorder resulting in motor dysfunction and cognitive decline. The inflammatory and neurodegenerative changes seen in the brains of MS patients lead to progressive disability and increasing brain atrophy. The most common type of MS is characterized by episodes of clinical exacerbations and remissions. This suggests the presence of compensating mechanisms for accumulating damage. Apart from the widely known repair mechanisms like remyelination, another important phenomenon is neuronal plasticity. Initially, neuroplasticity was connected with the developmental stages of life; however, there is now growing evidence confirming that structural and functional reorganization occurs throughout our lifetime. Several functional studies, utilizing such techniques as fMRI, TBS, or MRS, have provided valuable data about the presence of neuronal plasticity in MS patients. CNS ability to compensate for neuronal damage is most evident in RR-MS; however it has been shown that brain plasticity is also preserved in patients with substantial brain damage. Regardless of the numerous studies, the molecular background of neuronal plasticity in MS is still not well understood. Several factors, like IL-1β, BDNF, PDGF, or CB1Rs, have been implicated in functional recovery from the acute phase of MS and are thus considered as potential therapeutic targets.
Collapse
|
35
|
Blennow K, Mattsson N, Schöll M, Hansson O, Zetterberg H. Amyloid biomarkers in Alzheimer's disease. Trends Pharmacol Sci 2015; 36:297-309. [PMID: 25840462 DOI: 10.1016/j.tips.2015.03.002] [Citation(s) in RCA: 363] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/02/2015] [Accepted: 03/05/2015] [Indexed: 02/06/2023]
Abstract
Aggregation of amyloid-β (Aβ) into oligomers, fibrils, and plaques is central in the molecular pathogenesis of Alzheimer's disease (AD), and is the main focus of AD drug development. Biomarkers to monitor Aβ metabolism and aggregation directly in patients are important for further detailed study of the involvement of Aβ in disease pathogenesis and to monitor the biochemical effect of drugs targeting Aβ in clinical trials. Furthermore, if anti-Aβ disease-modifying drugs prove to be effective clinically, amyloid biomarkers will be of special value in the clinic to identify patients with brain amyloid deposition at risk for progression to AD dementia, to enable initiation of treatment before neurodegeneration is too severe, and to monitor drug effects on Aβ metabolism or pathology to guide dosage. Two types of amyloid biomarker have been developed: Aβ-binding ligands for use in positron emission tomography (PET) and assays to measure Aβ42 in cerebrospinal fluid (CSF). In this review, we present the rationales behind these biomarkers and compare their ability to measure Aβ plaque load in the brain. We also review possible shortcomings and the need of standardization of both biomarkers, as well as their implementation in the clinic.
Collapse
Affiliation(s)
- Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; The Torsten Söderberg Professorship at the Royal Swedish Academy of Sciences.
| | - Niklas Mattsson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Michael Schöll
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Department of Clinical Neuroscience and Rehabilitation, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences, Lund University, Lund, Sweden; Clinical Memory Research unit, Clinical Sciences, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
36
|
Cuchillo-Ibañez I, Lopez-Font I, Boix-Amorós A, Brinkmalm G, Blennow K, Molinuevo JL, Sáez-Valero J. Heteromers of amyloid precursor protein in cerebrospinal fluid. Mol Neurodegener 2015; 10:2. [PMID: 25573162 PMCID: PMC4298044 DOI: 10.1186/1750-1326-10-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/27/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Soluble fragments of the amyloid precursor protein (APP) generated by α- and β-secretases, sAPPα and sAPPβ, have been postulated as promising new cerebrospinal fluid (CSF) biomarkers for the clinical diagnosis of Alzheimer's disease (AD). However, the capacity of these soluble proteins to assemble has not been explored and could be relevant. Our aim is to characterize possible sAPP oligomers that could contribute to the quantification of sAPPα and sAPPβ in CSF by ELISA, as well as to characterize the possible presence of soluble full-length APP (sAPPf). RESULTS We employed co-immunoprecipitation, native polyacrylamide gel electrophoresis and ultracentrifugation in sucrose density gradients to characterize sAPP oligomers in CSF. We have characterized the presence of sAPPf in CSF from NDC and AD subjects and demonstrated that all forms, including sAPPα and sAPPβ, are capable of assembling into heteromers, which differ from brain APP membrane-dimers. We measured sAPPf, sAPPα and sAPPβ by ELISA in CSF samples from AD (n = 13) and non-disease subjects (NDC, n = 13) before and after immunoprecipitation with antibodies against the C-terminal APP or against sAPPβ. We demonstrated that these sAPP heteromers participate in the quantification of sAPPα and sAPPβ by ELISA. Immunoprecipitation with a C-terminal antibody to remove sAPPf reduced by ~30% the determinations of sAPPα and sAPPβ by ELISA, whereas immunoprecipitation with an APPβ antibody reduced by ~80% the determination of sAPPf and sAPPα. CONCLUSIONS The presence of sAPPf and sAPP heteromers should be taken into consideration when exploring the levels of sAPPα and sAPPβ as potential CSF biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Av, Ramón y Cajal s/n, Sant Joan d'Alacant, Spain.
| |
Collapse
|
37
|
Mattsson N, Insel PS, Donohue M, Landau S, Jagust WJ, Shaw LM, Trojanowski JQ, Zetterberg H, Blennow K, Weiner MW. Independent information from cerebrospinal fluid amyloid-β and florbetapir imaging in Alzheimer's disease. ACTA ACUST UNITED AC 2014; 138:772-83. [PMID: 25541191 DOI: 10.1093/brain/awu367] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Reduced cerebrospinal fluid amyloid-β42 and increased retention of florbetapir positron emission tomography are biomarkers reflecting cortical amyloid load in Alzheimer's disease. However, these measurements do not always agree and may represent partly different aspects of the underlying Alzheimer's disease pathology. The goal of this study was therefore to test if cerebrospinal fluid and positron emission tomography amyloid-β biomarkers are independently related to other Alzheimer's disease markers, and to examine individuals who are discordantly classified by these two biomarker modalities. Cerebrospinal fluid and positron emission tomography amyloid-β were measured at baseline in 769 persons [161 healthy controls, 68 subjective memory complaints, 419 mild cognitive impairment and 121 Alzheimer's disease dementia, mean age 72 years (standard deviation 7 years), 47% females] and used to predict diagnosis, APOE ε4 carriage status, cerebral blood flow, cerebrospinal fluid total-tau and phosphorylated-tau levels (cross-sectionally); and hippocampal volume, fluorodeoxyglucose positron emission tomography results and Alzheimer's Disease Assessment Scale-cognitive subscale scores (longitudinally). Cerebrospinal fluid and positron emission tomography amyloid-β were highly correlated, but adjusting one of these predictors for the other revealed that they both provided partially independent information when predicting diagnosis, APOE ε4, hippocampal volume, metabolism, cognition, total-tau and phosphorylated-tau (the 95% confidence intervals of the adjusted effects did not include zero). Cerebrospinal fluid amyloid-β was more strongly related to APOE ε4 whereas positron emission tomography amyloid-β was more strongly related to tau levels (P < 0.05). Discordance (mainly isolated cerebrospinal fluid amyloid-β positivity) differed by diagnostic group (P < 0.001) and was seen in 21% of cognitively healthy people but only 6% in dementia patients. The finding that cerebrospinal fluid and positron emission tomography amyloid-β provide partially independent information about a wide range of Alzheimer's measures supports the theory that these modalities represent partly different aspects of Alzheimer's pathology. The fact that mismatch, with positive cerebrospinal fluid amyloid-β but normal positron emission tomography amyloid-β, is relatively common in cognitively healthy people may be considered when using these biomarkers to identify early stage Alzheimer's disease. Reduced cerebrospinal fluid amyloid-β may be more strongly related to early stage Alzheimer's disease, whereas increased positron emission tomography amyloid-β may be more strongly related to disease progression.
Collapse
Affiliation(s)
- Niklas Mattsson
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden 2 Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA 3 Department of Veterans Affairs Medical Centre, Centre for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Philip S Insel
- 2 Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA 3 Department of Veterans Affairs Medical Centre, Centre for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Michael Donohue
- 4 Division of Biostatistics and Bioinformatics, Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Susan Landau
- 5 Helen Wills Neuroscience Institute and School of Public Health, University of California, Berkeley, CA, USA
| | - William J Jagust
- 5 Helen Wills Neuroscience Institute and School of Public Health, University of California, Berkeley, CA, USA
| | - Leslie M Shaw
- 6 Department of Pathology and Laboratory Medicine, Institute on Aging, Centre for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John Q Trojanowski
- 6 Department of Pathology and Laboratory Medicine, Institute on Aging, Centre for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Henrik Zetterberg
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden 7 UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Kaj Blennow
- 1 Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael W Weiner
- 2 Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA 3 Department of Veterans Affairs Medical Centre, Centre for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | | |
Collapse
|
38
|
Mattsson N, Insel PS, Landau S, Jagust W, Donohue M, Shaw LM, Trojanowski JQ, Zetterberg H, Blennow K, Weiner M. Diagnostic accuracy of CSF Ab42 and florbetapir PET for Alzheimer's disease. Ann Clin Transl Neurol 2014; 1:534-43. [PMID: 25356425 PMCID: PMC4184556 DOI: 10.1002/acn3.81] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/13/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Reduced cerebrospinal fluid (CSF) β-amyloid42 (Aβ42) and increased florbetapir positron emission tomography (PET) uptake reflects brain Aβ accumulation. These biomarkers are correlated with each other and altered in Alzheimer's disease (AD), but no study has directly compared their diagnostic performance. METHODS We examined healthy controls (CN, N = 169) versus AD dementia patients (N = 118), and stable (sMCI; no dementia, followed up for at least 2 years, N = 165) versus progressive MCI (pMCI; conversion to AD dementia, N = 59). All subjects had florbetapir PET (global and regional; temporal, frontal, parietal, and cingulate) and CSF Aβ42 measurements at baseline. We compared area under the curve (AUC), sensitivity, and specificity (testing a priori and optimized cutoffs). Clinical diagnosis was the reference standard. RESULTS CSF Aβ42 and (global or regional) PET florbetapir did not differ in AUC (CN vs. AD, CSF 84.4%; global PET 86.9%; difference [95% confidence interval] -6.7 to 1.5). CSF Aβ42 and global PET florbetapir did not differ in sensitivity, but PET had greater specificity than CSF in most comparisons. Sixteen CN progressed to MCI and AD (six Aβ negative, seven Aβ positive, and three PET positive but CSF negative). INTERPRETATION The overall diagnostic accuracies of CSF Aβ42 and PET florbetapir were similar, but PET had greater specificity. This was because some CN and sMCI subjects appear pathological using CSF but not using PET, suggesting that low CSF Aβ42 not always translates to cognitive decline or brain Aβ accumulation. Other factors, including costs and side effects, may also be considered when determining the optimal modality for different applications.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg Mölndal, Sweden ; Department of Radiology and Biomedical Imaging, University of California San Francisco, California ; Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases San Francisco, California
| | - Philip S Insel
- Department of Radiology and Biomedical Imaging, University of California San Francisco, California
| | - Susan Landau
- Helen Wills Neuroscience Institute and School of Public Health, University of California Berkeley, California
| | - William Jagust
- Helen Wills Neuroscience Institute and School of Public Health, University of California Berkeley, California
| | - Michael Donohue
- Division of Biostatistics and Bioinformatics, Department of Family & Preventive Medicine, University of California, San Diego La Jolla, California
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine Philadelphia, Pennsylvania
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging, Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine Philadelphia, Pennsylvania
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg Mölndal, Sweden ; UCL Institute of Neurology Queen Square, London, WC1N 3BG, United Kingdom
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg Mölndal, Sweden
| | - Michael Weiner
- Department of Radiology and Biomedical Imaging, University of California San Francisco, California ; Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases San Francisco, California
| | | |
Collapse
|
39
|
Affiliation(s)
- Lawrence Steinman
- Departments of Pediatrics, Neurology and Neurological Sciences, Stanford University, Stanford, California 94305;
| |
Collapse
|
40
|
Cerebrospinal Fluid Biomarkers in Alzheimer’s Disease and Frontotemporal Dementia. NEURODEGENER DIS 2014. [DOI: 10.1007/978-1-4471-6380-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
41
|
Lista S, Garaci FG, Ewers M, Teipel S, Zetterberg H, Blennow K, Hampel H. CSF Aβ1-42 combined with neuroimaging biomarkers in the early detection, diagnosis and prediction of Alzheimer's disease. Alzheimers Dement 2013; 10:381-92. [PMID: 23850330 DOI: 10.1016/j.jalz.2013.04.506] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 04/29/2013] [Indexed: 11/17/2022]
Abstract
The development of validated, qualified, and standardized biomarkers for Alzheimer's disease (AD) that allow for an early presymptomatic diagnosis and discrimination (classification) from other types of dementia and neurodegenerative diseases is warranted to accelerate the successful development of novel disease-modifying therapies. Here, we focus on the value of the 42-residue-long amyloid β isoform (Aβ1-42) peptide in the cerebrospinal fluid as the core, feasible neurobiochemical marker for the amyloidogenic mechanisms in early-onset familial and late-onset sporadic AD. We discuss the role and use of Aβ1-42 in combination with evolving neuroimaging biomarkers in AD detection and diagnosis. Multimodal neuroimaging techniques, directly providing structural-functional-metabolic aspects of brain pathophysiology, are supportive to predict and monitor the progression of the disease. Advances in multimodal neuroimaging provide new insights into brain organization and enable the detection of specific proteins and/or protein aggregates associated with AD. The combination of biomarkers from different methodologies is believed to be of incrementally added risk-value to accurately identify asymptomatic and prodromal individuals who will likely progress to dementia and represent rational biomarker candidates for preventive and symptomatic pharmacological intervention trials.
Collapse
Affiliation(s)
- Simone Lista
- Department of Psychiatry, Goethe-University, Frankfurt am Main, Germany.
| | - Francesco G Garaci
- Department of Diagnostic Imaging, Molecular Imaging, Interventional Radiology, and Radiotherapy, University of Rome "Tor Vergata," Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy
| | - Michael Ewers
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Stefan Teipel
- Department of Psychiatry, University of Rostock, Rostock, Germany DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; University College London Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Harald Hampel
- Department of Psychiatry, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|