51
|
Duran-Aniotz C, Orellana P, Leon Rodriguez T, Henriquez F, Cabello V, Aguirre-Pinto MF, Escobedo T, Takada LT, Pina-Escudero SD, Lopez O, Yokoyama JS, Ibanez A, Parra MA, Slachevsky A. Systematic Review: Genetic, Neuroimaging, and Fluids Biomarkers for Frontotemporal Dementia Across Latin America Countries. Front Neurol 2021; 12:663407. [PMID: 34248820 PMCID: PMC8263937 DOI: 10.3389/fneur.2021.663407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Frontotemporal dementia (FTD) includes a group of clinically, genetically, and pathologically heterogeneous neurodegenerative disorders, affecting the fronto-insular-temporal regions of the brain. Clinically, FTD is characterized by progressive deficits in behavior, executive function, and language and its diagnosis relies mainly on the clinical expertise of the physician/consensus group and the use of neuropsychological tests and/or structural/functional neuroimaging, depending on local availability. The modest correlation between clinical findings and FTD neuropathology makes the diagnosis difficult using clinical criteria and often leads to underdiagnosis or misdiagnosis, primarily due to lack of recognition or awareness of FTD as a disease and symptom overlap with psychiatric disorders. Despite advances in understanding the underlying neuropathology of FTD, accurate and sensitive diagnosis for this disease is still lacking. One of the major challenges is to improve diagnosis in FTD patients as early as possible. In this context, biomarkers have emerged as useful methods to provide and/or complement clinical diagnosis for this complex syndrome, although more evidence is needed to incorporate most of them into clinical practice. However, most biomarker studies have been performed using North American or European populations, with little representation of the Latin American and the Caribbean (LAC) region. In the LAC region, there are additional challenges, particularly the lack of awareness and knowledge about FTD, even in specialists. Also, LAC genetic heritage and cultures are complex, and both likely influence clinical presentations and may modify baseline biomarker levels. Even more, due to diagnostic delay, the clinical presentation might be further complicated by both neurological and psychiatric comorbidity, such as vascular brain damage, substance abuse, mood disorders, among others. This systematic review provides a brief update and an overview of the current knowledge on genetic, neuroimaging, and fluid biomarkers for FTD in LAC countries. Our review highlights the need for extensive research on biomarkers in FTD in LAC to contribute to a more comprehensive understanding of the disease and its associated biomarkers. Dementia research is certainly reduced in the LAC region, highlighting an urgent need for harmonized, innovative, and cross-regional studies with a global perspective across multiple areas of dementia knowledge.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Paulina Orellana
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Tomas Leon Rodriguez
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Henriquez
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Victoria Cabello
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Tamara Escobedo
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Leonel T. Takada
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
| | - Stefanie D. Pina-Escudero
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Oscar Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer S. Yokoyama
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, & National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Mario A. Parra
- School of Psychological Sciences and Health, University of Strathclyde, Glasgow, United Kingdom
| | - Andrea Slachevsky
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
- Department of Neurology and Psychiatry, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
52
|
Non-neuronal cells in amyotrophic lateral sclerosis - from pathogenesis to biomarkers. Nat Rev Neurol 2021; 17:333-348. [PMID: 33927394 DOI: 10.1038/s41582-021-00487-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 02/04/2023]
Abstract
The prevailing motor neuron-centric view of amyotrophic lateral sclerosis (ALS) pathogenesis could be an important factor in the failure to identify disease-modifying therapy for this neurodegenerative disorder. Non-neuronal cells have crucial homeostatic functions within the CNS and evidence of involvement of these cells in the pathophysiology of several neurodegenerative disorders, including ALS, is accumulating. Microglia and astrocytes, in crosstalk with peripheral immune cells, can exert both neuroprotective and adverse effects, resulting in a highly nuanced range of neuronal and non-neuronal cell interactions. This Review provides an overview of the diverse roles of non-neuronal cells in relation to the pathogenesis of ALS and the emerging potential of non-neuronal cell biomarkers to advance therapeutic development.
Collapse
|
53
|
Gaur N, Huss E, Prell T, Steinbach R, Guerra J, Srivastava A, Witte OW, Grosskreutz J. Monocyte-Derived Macrophages Contribute to Chitinase Dysregulation in Amyotrophic Lateral Sclerosis: A Pilot Study. Front Neurol 2021; 12:629332. [PMID: 34054686 PMCID: PMC8160083 DOI: 10.3389/fneur.2021.629332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/21/2021] [Indexed: 12/01/2022] Open
Abstract
Neuroinflammation significantly contributes to Amyotrophic Lateral Sclerosis (ALS) pathology. In lieu of this, reports of elevated chitinase levels in ALS are interesting, as they are established surrogate markers of a chronic inflammatory response. While post-mortem studies have indicated glial expression, the cellular sources for these moieties remain to be fully understood. Therefore, the objective of this pilot study was to examine whether the peripheral immune system also contributes to chitinase dysregulation in ALS. The temporal expression of CHIT1, CHI3L1, and CHI3L2 in non-polarized monocyte-derived macrophages (MoMas) from ALS patients and healthy controls (HCs) was examined. We demonstrate that while CHIT1 and CHI3L1 display similar temporal expression dynamics in both groups, profound between-group differences were noted for these targets at later time-points i.e., when cells were fully differentiated. CHIT1 and CHI3L1 expression were significantly higher in MoMas from ALS patients at both the transcriptomic and protein level, with CHI3L1 levels also being influenced by age. Conversely, CHI3L2 expression was not influenced by disease state, culture duration, or age. Here, we demonstrate for the first time, that in ALS, circulating immune cells have an intrinsically augmented potential for chitinase production that may propagate chronic neuroinflammation, and how the ageing immune system itself contributes to neurodegeneration.
Collapse
Affiliation(s)
- Nayana Gaur
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Elena Huss
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Tino Prell
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| | - Robert Steinbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Joel Guerra
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Akash Srivastava
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| |
Collapse
|
54
|
Mahoney CJ, Ahmed RM, Huynh W, Tu S, Rohrer JD, Bedlack RS, Hardiman O, Kiernan MC. Pathophysiology and Treatment of Non-motor Dysfunction in Amyotrophic Lateral Sclerosis. CNS Drugs 2021; 35:483-505. [PMID: 33993457 DOI: 10.1007/s40263-021-00820-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal neurodegenerative disease typically presenting with bulbar or limb weakness. There is increasing evidence that amyotrophic lateral sclerosis is a multisystem disease with early and frequent impacts on cognition, behaviour, sleep, pain and fatigue. Dysfunction of normal physiological and metabolic processes also appears common. Evidence from pre-symptomatic studies and large epidemiological cohorts examining risk factors for the future development of amyotrophic lateral sclerosis have reported a high prevalence of changes in behaviour and mental health before the emergence of motor weakness. This suggests that changes beyond the motor system are underway at an early stage with dysfunction across brain networks regulating a variety of cognitive, behavioural and other homeostatic processes. The full impact of non-motor dysfunction continues to be established but there is now sufficient evidence that the presence of non-motor symptoms impacts overall survival in amyotrophic lateral sclerosis, and with up to 80% reporting non-motor symptoms, there is an urgent need to develop more robust therapeutic approaches. This review provides a contemporary overview of the pathobiology of non-motor dysfunction, offering readers a practical approach with regard to assessment and management. We review the current evidence for pharmacological and non-pharmacological treatment of non-motor dysfunction in amyotrophic lateral sclerosis and highlight the need to further integrate non-motor dysfunction as an important outcome measure for future clinical trial design.
Collapse
Affiliation(s)
- Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Rebekah M Ahmed
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.,Department of Neurology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Richard S Bedlack
- Department of Neurology, Duke University Hospital, Durham, North Carolina, USA
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.,Department of Neurology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
55
|
van der Ende EL, Morenas-Rodriguez E, McMillan C, Grossman M, Irwin D, Sanchez-Valle R, Graff C, Vandenberghe R, Pijnenburg YAL, Laforce R, Ber IL, Lleo A, Haass C, Suarez-Calvet M, van Swieten JC, Seelaar H. CSF sTREM2 is elevated in a subset in GRN-related frontotemporal dementia. Neurobiol Aging 2021; 103:158.e1-158.e5. [PMID: 33896652 DOI: 10.1016/j.neurobiolaging.2021.02.024] [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: 12/18/2020] [Accepted: 02/27/2021] [Indexed: 02/06/2023]
Abstract
Excessive microglial activation might be a central pathological process in GRN-related frontotemporal dementia (FTD-GRN). We measured soluble triggering receptor expressed on myeloid cells 2 (sTREM2), which is shed from disease-associated microglia following cleavage of TREM2, in cerebrospinal fluid of 34 presymptomatic and 35 symptomatic GRN mutation carriers, 6 presymptomatic and 32 symptomatic C9orf72 mutation carriers and 67 healthy noncarriers by ELISA. Although no group differences in sTREM2 levels were observed (GRN: symptomatic (median 5.2 ng/mL, interquartile range [3.9-9.2]) vs. presymptomatic (4.3 ng/mL [2.6-6.1]) vs. noncarriers (4.2 ng/mL [2.6-5.5]): p = 0.059; C9orf72: symptomatic (4.3 [2.9-7.0]) vs. presymptomatic (3.2 [2.2-4.2]) vs. noncarriers: p = 0.294), high levels were seen in a subset of GRN, but not C9orf72, mutation carriers, which might reflect differential TREM2-related microglial activation. Interestingly, 2 presymptomatic carriers with low sTREM2 levels developed symptoms after 1 year, whereas 2 with high levels became symptomatic after >5 years. While sTREM2 is not a promising diagnostic biomarker for FTD-GRN or FTD-C9orf72, further research might elucidate its potential to monitor microglial activity and predict disease progression.
Collapse
Affiliation(s)
- Emma L van der Ende
- Alzheimer Center Rotterdam and Dept. of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Estrella Morenas-Rodriguez
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximillians-Universität München, Munich, Germany
| | - Corey McMillan
- Dept. of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Murray Grossman
- Dept. of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Irwin
- Dept. of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clinic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Caroline Graff
- Karolinska Institutet, Dept. NVS, Division of Neurogeriatrics, Bioclinicum, Stockholm, Sweden; Unit of Hereditary Dementia, Theme Aging, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire du CHU de Québec, Département des Sciences Neurologiques, Université Laval, Québec, Canada
| | - Isabelle Le Ber
- APHP, Reference Centre for Rare or Early Onset Dementias, IM2A, Department of Neurology, Hôpital La Pitié-Salpêtrière, Paris, France; Sorbonne Université, Paris Brain Institute, Institut du Cerveau, ICM, Inserm U1127, CNRS UMR 7225, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alberto Lleo
- Neurology Department, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximillians-Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marc Suarez-Calvet
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - John C van Swieten
- Alzheimer Center Rotterdam and Dept. of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Harro Seelaar
- Alzheimer Center Rotterdam and Dept. of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
56
|
Steinacker P, Feneberg E, Halbgebauer S, Witzel S, Verde F, Oeckl P, Van Damme P, Gaur N, Gray E, Grosskreutz J, Jardel CG, Kachanov M, Kuhle J, Lamari F, Maceski A, Del Mar Amador M, Mayer B, Morelli C, Petri S, Poesen K, Raaphorst J, Salachas F, Silani V, Turner MR, Verbeek MM, Volk AE, Weishaupt JH, Weydt P, Ludolph AC, Otto M. Chitotriosidase as biomarker for early stage amyotrophic lateral sclerosis: a multicenter study. Amyotroph Lateral Scler Frontotemporal Degener 2021; 22:276-286. [PMID: 33576252 DOI: 10.1080/21678421.2020.1861023] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Objective: Levels of chitotriosidase (CHIT1) are increased in the cerebrospinal fluid (CSF) of amyotrophic lateral sclerosis (ALS) patients reflecting microglial activation. Here, we determine the diagnostic and prognostic potential of CHIT1 for early symptomatic ALS. Methods: Overall, 275 patients from 8 European neurological centers were examined. We included ALS with <6 and >6 months from symptom onset, other motoneuron diseases (oMND), ALS mimics (DCon) and non-neurodegenerative controls (Con). CSF CHIT1 levels were analyzed for diagnostic power and association with progression and survival in comparison to the benchmark neurofilament. The 24-bp duplication polymorphism of CHIT1 was analyzed in a subset of patients (N = 65). Results: Homozygous CHIT1 duplication mutation carriers (9%) invariably had undetectable CSF CHIT1 levels, while heterozygous carriers had similar levels as patients with wildtype CHIT1 (p = 0.414). In both early and late symptomatic ALS CHIT1 levels was increased, did not correlate with patients' progression rates, and was higher in patients diagnosed with higher diagnostic certainty. Neurofilament levels correlated with CHIT1 levels and prevailed over CHIT1 regarding diagnostic performance. Both CHIT1 and neurofilaments were identified as independent predictors of survival in late but not early symptomatic ALS. Evidence is provided that CHIT1 predicts progression in El Escorial diagnostic category in the group of ALS cases with a short duration. Conclusions: CSF CHIT1 level may have additional value in the prognostication of ALS patients with a short history of symptoms classified in diagnostic categories of lower clinical certainty. To fully interpret apparently low CHIT1 levels knowledge of CHIT1 genotype is needed.
Collapse
Affiliation(s)
| | - Emily Feneberg
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Simon Witzel
- Department of Neurology, Ulm University, Ulm, Germany
| | - Federico Verde
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Pathophysiology and Transplantation - "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Patrick Oeckl
- Department of Neurology, Ulm University, Ulm, Germany
| | - Philip Van Damme
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Department of Neurosciences, VIB - Center for Brain & Disease Research, Experimental Neurology - Laboratory of Neurobiology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Nayana Gaur
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Elizabeth Gray
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Claude G Jardel
- Department of Metabolic Biochemistry, Hôpitaux Universitaires Pitié Salpeêtrière-Charles Foix, Paris, France
| | - Mykyta Kachanov
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Kuhle
- Neurology, Departments of Biomedicine, Medicine and Clinical Research, University and University Hospital Basel, Basel, Switzerland
| | - Foudil Lamari
- Department of Metabolic Biochemistry, Hôpitaux Universitaires Pitié Salpeêtrière-Charles Foix, Paris, France
| | - Aleksandra Maceski
- Neurology, Departments of Biomedicine, Medicine and Clinical Research, University and University Hospital Basel, Basel, Switzerland
| | - Maria Del Mar Amador
- Neurological Diseases Department, Paris ALS Reference Center, Hôpitaux Universitaires Pitié Salpeêtrière-Charles Foix, Paris, France
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Claudia Morelli
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Koen Poesen
- Laboratory of Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven and Laboratory Medicine, University Hospitals of Leuven, Leuven, Belgium
| | - Joost Raaphorst
- Department of Neurology, Amsterdam Neuroscience Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - François Salachas
- Neurological Diseases Department, Paris ALS Reference Center, Hôpitaux Universitaires Pitié Salpeêtrière-Charles Foix, Paris, France
| | - Vincenzo Silani
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Pathophysiology and Transplantation - "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Marcel M Verbeek
- Departments of Neurology and Laboratory Medicine, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander E Volk
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Patrick Weydt
- Department for Neurodegenerative Disorders and Gerontopsychiatry, Bonn University, Bonn, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
| |
Collapse
|
57
|
Swift IJ, Sogorb-Esteve A, Heller C, Synofzik M, Otto M, Graff C, Galimberti D, Todd E, Heslegrave AJ, van der Ende EL, Van Swieten JC, Zetterberg H, Rohrer JD. Fluid biomarkers in frontotemporal dementia: past, present and future. J Neurol Neurosurg Psychiatry 2021; 92:204-215. [PMID: 33188134 DOI: 10.1136/jnnp-2020-323520] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/03/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022]
Abstract
The frontotemporal dementia (FTD) spectrum of neurodegenerative disorders includes a heterogeneous group of conditions. However, following on from a series of important molecular studies in the early 2000s, major advances have now been made in the understanding of the pathological and genetic underpinnings of the disease. In turn, alongside the development of novel methodologies for measuring proteins and other molecules in biological fluids, the last 10 years have seen a huge increase in biomarker studies within FTD. This recent past has focused on attempting to develop markers that will help differentiate FTD from other dementias (particularly Alzheimer's disease (AD)), as well as from non-neurodegenerative conditions such as primary psychiatric disorders. While cerebrospinal fluid, and more recently blood, markers of AD have been successfully developed, specific markers identifying primary tauopathies or TDP-43 proteinopathies are still lacking. More focus at the moment has been on non-specific markers of neurodegeneration, and in particular, multiple studies of neurofilament light chain have highlighted its importance as a diagnostic, prognostic and staging marker of FTD. As clinical trials get under way in specific genetic forms of FTD, measures of progranulin and dipeptide repeat proteins in biofluids have become important potential measures of therapeutic response. However, understanding of whether drugs restore cellular function will also be important, and studies of key pathophysiological processes, including neuroinflammation, lysosomal function and synaptic health, are also now becoming more common. There is much still to learn in the fluid biomarker field in FTD, but the creation of large multinational cohorts is facilitating better powered studies and will pave the way for larger omics studies, including proteomics, metabolomics and lipidomics, as well as investigations of multimodal biomarker combinations across fluids, brain imaging and other domains. Here we provide an overview of the past, present and future of fluid biomarkers within the FTD field.
Collapse
Affiliation(s)
- Imogen Joanna Swift
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK.,Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Carolin Heller
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Caroline Graff
- Division for Neurogeriatrics, Center for Alzheimer Research, Department of NVS, Karolinska Institutet, Stockholm, Sweden.,Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan, Italy.,Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Emily Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Amanda J Heslegrave
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
| | | | | | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan Daniel Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
58
|
Ghasemi M, Keyhanian K, Douthwright C. Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells 2021; 10:cells10020249. [PMID: 33525344 PMCID: PMC7912327 DOI: 10.3390/cells10020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of the chromosome 9 open reading frame 72 (C9orf72) repeat expansion mutation in 2011 as the most common genetic abnormality in amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's disease) and frontotemporal dementia (FTD), progress in understanding the signaling pathways related to this mutation can only be described as intriguing. Two major theories have been suggested-(i) loss of function or haploinsufficiency and (ii) toxic gain of function from either C9orf72 repeat RNA or dipeptide repeat proteins (DPRs) generated from repeat-associated non-ATG (RAN) translation. Each theory has provided various signaling pathways that potentially participate in the disease progression. Dysregulation of the immune system, particularly glial cell dysfunction (mainly microglia and astrocytes), is demonstrated to play a pivotal role in both loss and gain of function theories of C9orf72 pathogenesis. In this review, we discuss the pathogenic roles of glial cells in C9orf72 ALS/FTD as evidenced by pre-clinical and clinical studies showing the presence of gliosis in C9orf72 ALS/FTD, pathologic hallmarks in glial cells, including TAR DNA-binding protein 43 (TDP-43) and p62 aggregates, and toxicity of C9orf72 glial cells. A better understanding of these pathways can provide new insights into the development of therapies targeting glial cell abnormalities in C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Correspondence: ; Tel.: +1-774-441-7726; Fax: +1-508-856-4485
| | | | | |
Collapse
|
59
|
Fluid Biomarkers of Frontotemporal Lobar Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:123-139. [PMID: 33433873 DOI: 10.1007/978-3-030-51140-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A timely diagnosis of frontotemporal degeneration (FTD) is frequently challenging due to the heterogeneous symptomatology and poor phenotype-pathological correlation. Fluid biomarkers that reflect FTD pathophysiology could be instrumental in both clinical practice and pharmaceutical trials. In recent years, significant progress has been made in developing biomarkers of neurodegenerative diseases: amyloid-β and tau in cerebrospinal fluid (CSF) can be used to exclude Alzheimer's disease, while neurofilament light chain (NfL) is emerging as a promising, albeit nonspecific, marker of neurodegeneration in both CSF and blood. Gene-specific biomarkers such as PGRN in GRN mutation carriers and dipeptide repeat proteins in C9orf72 mutation carriers are potential target engagement markers in genetic FTD trials. Novel techniques capable of measuring very low concentrations of brain-derived proteins in peripheral fluids are facilitating studies of blood biomarkers as a minimally invasive alternative to CSF. A major remaining challenge is the identification of a biomarker that can be used to predict the neuropathological substrate in sporadic FTD patients.
Collapse
|
60
|
Petrosino S, Schiano Moriello A. Palmitoylethanolamide: A Nutritional Approach to Keep Neuroinflammation within Physiological Boundaries-A Systematic Review. Int J Mol Sci 2020; 21:E9526. [PMID: 33333772 PMCID: PMC7765232 DOI: 10.3390/ijms21249526] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Neuroinflammation is a physiological response aimed at maintaining the homodynamic balance and providing the body with the fundamental resource of adaptation to endogenous and exogenous stimuli. Although the response is initiated with protective purposes, the effect may be detrimental when not regulated. The physiological control of neuroinflammation is mainly achieved via regulatory mechanisms performed by particular cells of the immune system intimately associated with or within the nervous system and named "non-neuronal cells." In particular, mast cells (within the central nervous system and in the periphery) and microglia (at spinal and supraspinal level) are involved in this control, through a close functional relationship between them and neurons (either centrally, spinal, or peripherally located). Accordingly, neuroinflammation becomes a worsening factor in many disorders whenever the non-neuronal cell supervision is inadequate. It has been shown that the regulation of non-neuronal cells-and therefore the control of neuroinflammation-depends on the local "on demand" synthesis of the endogenous lipid amide Palmitoylethanolamide and related endocannabinoids. When the balance between synthesis and degradation of this bioactive lipid mediator is disrupted in favor of reduced synthesis and/or increased degradation, the behavior of non-neuronal cells may not be appropriately regulated and neuroinflammation exceeds the physiological boundaries. In these conditions, it has been demonstrated that the increase of endogenous Palmitoylethanolamide-either by decreasing its degradation or exogenous administration-is able to keep neuroinflammation within its physiological limits. In this review the large number of studies on the benefits derived from oral administration of micronized and highly bioavailable forms of Palmitoylethanolamide is discussed, with special reference to neuroinflammatory disorders.
Collapse
Affiliation(s)
- Stefania Petrosino
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy;
- Epitech Group SpA, Via Einaudi 13, 35030 Padova, Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Napoli, Italy;
- Epitech Group SpA, Via Einaudi 13, 35030 Padova, Italy
| |
Collapse
|
61
|
Benussi A, Ashton NJ, Karikari TK, Gazzina S, Premi E, Benussi L, Ghidoni R, Rodriguez JL, Emeršič A, Binetti G, Fostinelli S, Giunta M, Gasparotti R, Zetterberg H, Blennow K, Borroni B. Serum Glial Fibrillary Acidic Protein (GFAP) Is a Marker of Disease Severity in Frontotemporal Lobar Degeneration. J Alzheimers Dis 2020; 77:1129-1141. [DOI: 10.3233/jad-200608] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: It is still unknown if serum glial fibrillary acidic protein (GFAP) is a useful marker in frontotemporal lobar degeneration (FTLD). Objective: To assess the diagnostic and prognostic value of serum GFAP in a large cohort of patients with FTLD. Methods: In this retrospective study, performed on 406 participants, we measured serum GFAP concentration with an ultrasensitive Single molecule array (Simoa) method in patients with FTLD, Alzheimer’s disease (AD), and in cognitively unimpaired elderly controls. We assessed the role of GFAP as marker of disease severity by analyzing the correlation with clinical variables, neurophysiological data, and cross-sectional brain imaging. Moreover, we evaluated the role of serum GFAP as a prognostic marker of disease survival. Results: We observed significantly higher levels of serum GFAP in patients with FTLD syndromes, except progressive supranuclear palsy, compared with healthy controls, but not compared with AD patients. In FTLD, serum GFAP levels correlated with measures of cognitive dysfunction and disease severity, and were associated with indirect measures of GABAergic deficit. Serum GFAP concentration was not a significant predictor of survival. Conclusion: Serum GFAP is increased in FTLD, correlates with cognition and GABAergic deficits, and thus shows promise as a biomarker of disease severity in FTLD.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Thomas K. Karikari
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | | | - Enrico Premi
- Stroke Unit, ASST Spedali Civili, Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Juan Lantero Rodriguez
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andreja Emeršič
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurology, University Medical Centre Ljubljana, Slovenia
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurology, University Medical Centre Ljubljana, Slovenia
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurology, University Medical Centre Ljubljana, Slovenia
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
62
|
Chen S, Liao Q, Lu K, Zhou J, Huang C, Bi F. Riluzole Exhibits No Therapeutic Efficacy on a Transgenic Rat model of Amyotrophic Lateral Sclerosis. Curr Neurovasc Res 2020; 17:275-285. [PMID: 32271694 DOI: 10.2174/1567202617666200409125227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurological disorder clinically characterized by motor system dysfunction, with intraneuronal accumulation of the TAR DNAbinding protein 43 (TDP-43) being a pathological hallmark. Riluzole is a primarily prescribed medicine for ALS patients, while its therapeutical efficacy appears limited. TDP-43 transgenic mice are existing animal models for mechanistic/translational research into ALS. METHODS We developed a transgenic rat model of ALS expressing a mutant human TDP-43 transgene (TDP-43M337V) and evaluated the therapeutic effect of Riluzole on this model. Relative to control, rats with TDP-43M337V expression promoted by the neurofilament heavy subunit (NEF) gene or specifically in motor neurons promoted by the choline acetyltransferase (ChAT) gene showed progressive worsening of mobility and grip strength, along with loss of motor neurons, microglial activation, and intraneuronal accumulation of TDP-43 and ubiquitin aggregations in the spinal cord. RESULTS Compared to vehicle control, intragastric administration of Riluzole (30 mg/kg/d) did not mitigate the behavioral deficits nor alter the neuropathologies in the transgenics. CONCLUSION These findings indicate that transgenic rats recapitulate the basic neurological and neuropathological characteristics of human ALS, while Riluzole treatment can not halt the development of the behavioral and histopathological phenotypes in this new transgenic rodent model of ALS.
Collapse
Affiliation(s)
- Si Chen
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Qiao Liao
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Ke Lu
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Jinxia Zhou
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| | - Cao Huang
- Department of Pathology Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Fangfang Bi
- Department of Neurology, Central South University, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
63
|
Obrador E, Salvador R, López-Blanch R, Jihad-Jebbar A, Vallés SL, Estrela JM. Oxidative Stress, Neuroinflammation and Mitochondria in the Pathophysiology of Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2020; 9:E901. [PMID: 32971909 PMCID: PMC7555310 DOI: 10.3390/antiox9090901] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron (MN) disease. Its primary cause remains elusive, although a combination of different causal factors cannot be ruled out. There is no cure, and prognosis is poor. Most patients with ALS die due to disease-related complications, such as respiratory failure, within three years of diagnosis. While the underlying mechanisms are unclear, different cell types (microglia, astrocytes, macrophages and T cell subsets) appear to play key roles in the pathophysiology of the disease. Neuroinflammation and oxidative stress pave the way leading to neurodegeneration and MN death. ALS-associated mitochondrial dysfunction occurs at different levels, and these organelles are involved in the mechanism of MN death. Molecular and cellular interactions are presented here as a sequential cascade of events. Based on our present knowledge, the discussion leads to the idea that feasible therapeutic strategies should focus in interfering with the pathophysiology of the disease at different steps.
Collapse
Affiliation(s)
| | | | | | | | | | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 4601016 Valencia, Spain; (E.O.); (R.S.); (R.L.-B.); (A.J.-J.); (S.L.V.)
| |
Collapse
|
64
|
Woollacott IO, Nicholas JM, Heller C, Foiani MS, Moore KM, Russell LL, Paterson RW, Keshavan A, Schott JM, Warren JD, Heslegrave A, Zetterberg H, Rohrer JD. Cerebrospinal Fluid YKL-40 and Chitotriosidase Levels in Frontotemporal Dementia Vary by Clinical, Genetic and Pathological Subtype. Dement Geriatr Cogn Disord 2020; 49:56-76. [PMID: 32344399 PMCID: PMC7513620 DOI: 10.1159/000506282] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic glial dysfunction may contribute to the pathogenesis of frontotemporal dementia (FTD). Cerebrospinal fluid (CSF) levels of glia-derived proteins YKL-40 and chitotriosidase are increased in Alzheimer's disease (AD) but have not been explored in detail across the spectrum of FTD. METHODS We investigated whether CSF YKL-40 and chitotriosidase levels differed between FTD patients and controls, across different clinical and genetic subtypes of FTD, and between individuals with a clinical FTD syndrome due to AD versus non-AD (frontotemporal lobar degeneration, FTLD) pathology (based on CSF neurodegenerative biomarkers). Eighteen healthy controls and 64 people with FTD (behavioural variant FTD, n = 20; primary progressive aphasia [PPA], n = 44: nfvPPA, n = 16, svPPA, n = 11, lvPPA, n = 14, PPA-NOS, n = 3) were included. 10/64 had familial FTD, with mutations in GRN(n = 3), MAPT(n = 4), or C9orf72 (n = 3). 15/64 had neurodegenerative biomarkers consistent with AD pathology. Levels were measured by immunoassay and compared using multiple linear regressions. We also examined relationships of YKL-40 and chitotriosidase with CSF total tau (T-tau), phosphorylated tau 181 (P-tau) and β-amyloid 1-42 (Aβ42), with each other, and with age and disease du-ration. RESULTS CSF YKL-40 and chitotriosidase levels were higher in FTD, particularly lvPPA (both) and nfvPPA (YKL-40), compared with controls. GRN mutation carriers had higher levels of both proteins than controls and C9orf72 expansion carriers, and YKL-40 was higher in MAPT mutation carriers than controls. Individuals with underlying AD pathology had higher YKL-40 and chitotriosidase levels than both controls and those with likely FTLD pathology. CSF YKL-40 and chitotriosidase levels were variably associated with levels of T-tau, P-tau and Aβ42, and with each other, depending on clinical syndrome and underlying pathology. CSF YKL-40 but not chitotriosidase was associated with age, but not disease duration. CONCLUSION CSF YKL-40 and chitotriosidase levels are increased in individuals with clinical FTD syndromes, particularly due to AD pathology. In a preliminary analysis of genetic groups, levels of both proteins are found to be highly elevated in FTD due to GRN mutations, while YKL-40 is increased in individuals with MAPT mutations. As glia-derived protein levels generally correlate with T-tau and P-tau levels, they may reflect the glial response to neurodegeneration in FTLD.
Collapse
Affiliation(s)
- Ione O.C. Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jennifer M. Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Carolin Heller
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Martha S. Foiani
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Katrina M. Moore
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Lucy L. Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Ross W. Paterson
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jonathan M. Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jason D. Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Amanda Heslegrave
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Henrik Zetterberg
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom,*Dr. Jonathan D. Rohrer, Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London WC1N 3BG (UK),
| |
Collapse
|
65
|
Altered serum protein levels in frontotemporal dementia and amyotrophic lateral sclerosis indicate calcium and immunity dysregulation. Sci Rep 2020; 10:13741. [PMID: 32792518 PMCID: PMC7426269 DOI: 10.1038/s41598-020-70687-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are neurodegenerative diseases that are considered to be on the same disease spectrum because of overlapping genetic, pathological and clinical traits. Changes in serum proteins in FTD and ALS are poorly understood, and currently no definitive biomarkers exist for diagnosing or monitoring disease progression for either disease. Here we applied quantitative discovery proteomics to analyze protein changes in FTD (N = 72) and ALS (N = 28) patient serum compared to controls (N = 22). Twenty three proteins were significantly altered in FTD compared to controls (increased-APOL1, C3, CTSH, EIF5A, MYH2, S100A8, SUSD5, WDR1; decreased-C1S, C7, CILP2, COMP, CRTAC1, EFEMP1, FBLN1, GSN, HSPG2, IGHV1, ITIH2, PROS1, SHBG, UMOD, VASN) and 14 proteins were significantly altered in ALS compared to controls (increased-APOL1, CKM, CTSH, IGHG1, IGKC, MYH2; decreased-C7, COMP, CRTAC1, EFEMP1, FBLN1, GSN, HSPG2, SHBG). There was substantial overlap in the proteins that were altered in FTD and ALS. These results were validated using western blotting. Gene ontology tools were used to assess functional pathways potentially dysregulated in the two diseases, and calcium ion binding and innate immunity pathways were altered in both diseases. When put together, these results suggest significant overlap in pathophysiological peripheral changes in FTD and ALS. This study represents the first proteomics side-by-side comparison of serum changes in FTD and ALS, providing new insights into under-recognized perturbed pathways and an avenue for biomarker development for FTD and ALS.
Collapse
|
66
|
Béland LC, Markovinovic A, Jakovac H, De Marchi F, Bilic E, Mazzini L, Kriz J, Munitic I. Immunity in amyotrophic lateral sclerosis: blurred lines between excessive inflammation and inefficient immune responses. Brain Commun 2020; 2:fcaa124. [PMID: 33134918 PMCID: PMC7585698 DOI: 10.1093/braincomms/fcaa124] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.
Collapse
Affiliation(s)
| | - Andrea Markovinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Ervina Bilic
- Department of Neurology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Letizia Mazzini
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec G1J 2G3, Canada
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
67
|
Gray E, Thompson AG, Wuu J, Pelt J, Talbot K, Benatar M, Turner MR. CSF chitinases before and after symptom onset in amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2020; 7:1296-1306. [PMID: 32666680 PMCID: PMC7448184 DOI: 10.1002/acn3.51114] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Objective To evaluate the CSF levels of chitinase proteins during the presymptomatic and early symptomatic phases of amyotrophic lateral sclerosis (ALS). Methods CSF samples were obtained from 16 controls, 55 individuals at‐risk for ALS (including 18 carrying a mutation in C9ORF72, 33 in SOD1), 12 ALS patients, and 7 phenoconverters (individuals diagnosed with ALS during follow‐up). At‐risk individuals and phenoconverters were enrolled through the Pre‐fALS study, which includes individuals carrying an ALS‐associated gene mutation without disease manifestations at initial assessment. Longitudinal CSF collections, where possible, took place every 3‐12 months for ALS patients and every 1‐2 years for others. CSF levels of chitotriosidase 1 (CHIT1), chitinase‐3‐like protein 1 (CHI3L1, YKL‐40) and chitinase‐3‐like protein 2 (CHI3L2, YKL‐39) were measured by ELISA, along with CHIT1 activity. Longitudinal changes in at‐risk individuals and phenoconverters were fitted to linear mixed effects models. Results Slowly rising levels of CHIT1 were observed over time in the at‐risk individuals (slope 0.059 log10[CHIT1] per year, P < 0.001). Among phenoconverters, CHIT1 levels and activity rose more sharply (0.403 log10[CHIT1] per year, P = 0.005; 0.260 log10[CHIT1 activity] per year, P = 0.007). Individual levels of both CHI3L1 and CHI3L2 remained relatively stable over time in all participant groups. Interpretation The CHIT1 neuroinflammatory response is a feature of the late presymptomatic to early symptomatic phases of ALS. This study does not suggest a long prodrome of upregulated glial activity in ALS pathogenesis, but strengthens the place of CHIT1 as part of a panel of biomarkers to objectively assess the impact of immune‐modulatory therapeutic interventions in ALS.
Collapse
Affiliation(s)
- Elizabeth Gray
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Joanne Wuu
- Department of Neurology, University of Miami, Miami, Florida
| | - Joe Pelt
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michael Benatar
- Department of Neurology, University of Miami, Miami, Florida
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
68
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
69
|
Gaur N, Perner C, Witte OW, Grosskreutz J. The Chitinases as Biomarkers for Amyotrophic Lateral Sclerosis: Signals From the CNS and Beyond. Front Neurol 2020; 11:377. [PMID: 32536900 PMCID: PMC7267218 DOI: 10.3389/fneur.2020.00377] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset neurodegenerative condition, most widely characterized by the selective vulnerability of motor neurons and the poor life expectancy of afflicted patients. Limited disease-modifying therapies currently exist, which only further attests to the substantial heterogeneity associated with this disease. In addition to established prognostic factors like genetic background, site of onset, and age at onset, wide consensus on the role of neuroinflammation as a disease exacerbator and driver has been established. In lieu of this, the emerging literature on chitinases in ALS is particularly intriguing. Individual groups have reported substantially elevated chitotriosidase (CHIT1), chitinase-3-like-1 (CHI3L1), and chitinase-3-like-2 (CHI3L2) levels in the cerebrospinal, motor cortex, and spinal cord of ALS patients with multiple—and often conflicting—lines of evidence hinting at possible links to disease severity and progression. This mini-review, while not exhaustive, will aim to discuss current evidence on the involvement of key chitinases in ALS within the wider framework of other neurodegenerative conditions. Implications for understanding disease etiology, developing immunomodulatory therapies and biomarkers, and other translational opportunities will be considered.
Collapse
Affiliation(s)
- Nayana Gaur
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Caroline Perner
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, MA, United States
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Center for Healthy Ageing, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Center for Healthy Ageing, Jena University Hospital, Jena, Germany
| |
Collapse
|
70
|
Barschke P, Oeckl P, Steinacker P, Al Shweiki MR, Weishaupt JH, Landwehrmeyer GB, Anderl-Straub S, Weydt P, Diehl-Schmid J, Danek A, Kornhuber J, Schroeter ML, Prudlo J, Jahn H, Fassbender K, Lauer M, van der Ende EL, van Swieten JC, Volk AE, Ludolph AC, Otto M. Different CSF protein profiles in amyotrophic lateral sclerosis and frontotemporal dementia with C9orf72 hexanucleotide repeat expansion. J Neurol Neurosurg Psychiatry 2020; 91:503-511. [PMID: 32132225 DOI: 10.1136/jnnp-2019-322476] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVES The hexanucleotide repeat expansion in the C9orf72 gene is the most common mutation associated with amyotrophic lateral sclerosis (C9-ALS) and frontotemporal dementia (C9-FTD). Until now, it is unknown which factors define whether C9orf72 mutation carriers develop ALS or FTD. Our aim was to identify protein biomarker candidates in the cerebrospinal fluid (CSF) which differentiate between C9-ALS and C9-FTD and might be indicative for the outcome of the mutation. METHODS We compared the CSF proteome of 16 C9-ALS and 8 C9-FTD patients and 11 asymptomatic C9orf72 mutation carriers (CAR) by isobaric tags for relative and absolute quantitation. Eleven biomarker candidates were selected from the pool of differentially regulated proteins for further validation by multiple reaction monitoring and single-molecule array in a larger cohort (n=156). RESULTS In total, 2095 CSF proteins were identified and 236 proteins were significantly different in C9-ALS versus C9-FTD including neurofilament medium polypeptide (NEFM) and chitotriosidase-1 (CHIT1). Eight candidates were successfully validated including significantly increased ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL1) levels in C9-ALS compared with C9-FTD and controls and decreased neuronal pentraxin receptor (NPTXR) levels in C9-FTD versus CAR. CONCLUSIONS This study presents a deep proteomic CSF analysis of C9-ALS versus C9-FTD patients. As a proof of concept, we observed higher NEFM and CHIT1 CSF levels in C9-ALS. In addition, we also show clear upregulation of UCHL1 in C9-ALS and downregulation of NPTXR in C9-FTD. Significant differences in UCHL1 CSF levels may explain diverging ubiquitination and autophagy processes and NPTXR levels might reflect different synapses organisation processes.
Collapse
Affiliation(s)
- Peggy Barschke
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Patrick Oeckl
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Petra Steinacker
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | | | - Jochen H Weishaupt
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | | | | | - Patrick Weydt
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians Universität, Munich, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias L Schroeter
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Saxony, Germany.,Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Johannes Prudlo
- Department of Neurology, Rostock University Medical Center, German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Holger Jahn
- Clinic for Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Fassbender
- Department of Neurology, University of Saarland, Homburg, Germany
| | - Martin Lauer
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University of Würzburg, Würzburg, Germany
| | | | | | - Alexander E Volk
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Albert C Ludolph
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Baden-Württemberg, Germany
| | | |
Collapse
|
71
|
Oldoni E, Smets I, Mallants K, Vandebergh M, Van Horebeek L, Poesen K, Dupont P, Dubois B, Goris A. CHIT1 at Diagnosis Reflects Long-Term Multiple Sclerosis Disease Activity. Ann Neurol 2020; 87:633-645. [PMID: 31997416 PMCID: PMC7187166 DOI: 10.1002/ana.25691] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Evidence for a role of microglia in the pathogenesis of multiple sclerosis (MS) is growing. We investigated association of microglial markers at time of diagnostic lumbar puncture (LP) with different aspects of disease activity (relapses, disability, magnetic resonance imaging parameters) up to 6 years later in a cohort of 143 patients. METHODS In cerebrospinal fluid (CSF), we measured 3 macrophage and microglia-related proteins, chitotriosidase (CHIT1), chitinase-3-like protein 1 (CHI3L1 or YKL-40), and soluble triggering receptor expressed on myeloid cells 2 (sTREM2), as well as a marker of neuronal damage, neurofilament light chain (NfL), using enzyme-linked immunosorbent assay and electrochemiluminescence. We investigated the same microglia-related markers in publicly available RNA expression data from postmortem brain tissue. RESULTS CHIT1 levels at diagnostic LP correlated with 2 aspects of long-term disease activity after correction for multiple testing. First, CHIT1 increased with reduced tissue integrity in lesions at a median 3 years later (p = 9.6E-04). Second, CHIT1 reflected disease severity at a median 5 years later (p = 1.2E-04). Together with known clinical covariates, CHIT1 levels explained 12% and 27% of variance in these 2 measures, respectively, and were able to distinguish slow and fast disability progression (area under the curve = 85%). CHIT1 was the best discriminator of chronic active versus chronic inactive lesions and the only marker correlated with NfL (r = 0.3, p = 0.0019). Associations with disease activity were, however, independent of NfL. INTERPRETATION CHIT1 CSF levels measured during the diagnostic LP reflect microglial activation early on in MS and can be considered a valuable prognostic biomarker for future disease activity. ANN NEUROL 2020;87:633-645.
Collapse
Affiliation(s)
- Emanuela Oldoni
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Ide Smets
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
- Department of Neurology, University Hospitals LeuvenLeuvenBelgium
| | - Klara Mallants
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Marijne Vandebergh
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Lies Van Horebeek
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Koen Poesen
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
- KU Leuven ‐ Department of Neurosciences, Laboratory for Molecular Neurobiomarker ResearchLeuvenBelgium
| | - Patrick Dupont
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
- KU Leuven ‐ Department of Neurosciences, Laboratory for Cognitive NeurologyLeuvenBelgium
| | - Bénédicte Dubois
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
- Department of Neurology, University Hospitals LeuvenLeuvenBelgium
| | - An Goris
- KU Leuven ‐ Department of Neurosciences, Laboratory for NeuroimmunologyLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| |
Collapse
|
72
|
Vu L, An J, Kovalik T, Gendron T, Petrucelli L, Bowser R. Cross-sectional and longitudinal measures of chitinase proteins in amyotrophic lateral sclerosis and expression of CHI3L1 in activated astrocytes. J Neurol Neurosurg Psychiatry 2020; 91:350-358. [PMID: 31937582 PMCID: PMC7147184 DOI: 10.1136/jnnp-2019-321916] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Amyotrophic lateral sclerosis (ALS) is a complex disease with numerous pathological mechanisms resulting in a heterogeneous patient population. Using biomarkers for particular disease mechanisms may enrich a homogeneous subset of patients. In this study, we quantified chitotriosidase (Chit-1) and chitinase-3-like protein 1 (CHI3L1), markers of glial activation, in cerebrospinal fluid (CSF) and plasma and determined the cell types that express CHI3L1 in ALS. METHODS Immunoassays were used to quantify Chit-1, CHI3L1 and phosphorylated neurofilament heavy chain levels in longitudinal CSF and matching plasma samples from 118 patients with ALS, 17 disease controls (DCs), and 24 healthy controls (HCs). Immunostaining was performed to identify and quantify CHI3L1-positive cells in tissue sections from ALS, DCs and non-neurological DCs. RESULTS CSF Chit-1 exhibited increased levels in ALS as compared with DCs and HCs. CSF CHI3L1 levels were increased in ALS and DCs compared with HCs. No quantitative differences were noted in plasma for either chitinase. Patients with ALS with fast-progressing disease exhibited higher levels of CSF Chit-1 and CHI3L1 than patients with slow-progressing disease. Increased numbers of CHI3L1-positive cells were observed in postmortem ALS motor cortex as compared with controls, and these cells were identified as a subset of activated astrocytes located predominately in the white matter of the motor cortex and the spinal cord. CONCLUSIONS CSF Chit-1 and CHI3L1 are significantly increased in ALS, and CSF Chit-1 and CHI3L1 levels correlate to the rate of disease progression. CHI3L1 is expressed by a subset of activated astrocytes predominately located in white matter.
Collapse
Affiliation(s)
- Lucas Vu
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Jiyan An
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Tina Kovalik
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Tania Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Robert Bowser
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA .,Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| |
Collapse
|
73
|
Heller C, Foiani MS, Moore K, Convery R, Bocchetta M, Neason M, Cash DM, Thomas D, Greaves CV, Woollacott IO, Shafei R, Van Swieten JC, Moreno F, Sanchez-Valle R, Borroni B, Laforce R, Masellis M, Tartaglia MC, Graff C, Galimberti D, Rowe JB, Finger E, Synofzik M, Vandenberghe R, de Mendonca A, Tagliavini F, Santana I, Ducharme S, Butler CR, Gerhard A, Levin J, Danek A, Frisoni G, Sorbi S, Otto M, Heslegrave AJ, Zetterberg H, Rohrer JD. Plasma glial fibrillary acidic protein is raised in progranulin-associated frontotemporal dementia. J Neurol Neurosurg Psychiatry 2020; 91:263-270. [PMID: 31937580 DOI: 10.1136/jnnp-2019-321954] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND There are few validated fluid biomarkers in frontotemporal dementia (FTD). Glial fibrillary acidic protein (GFAP) is a measure of astrogliosis, a known pathological process of FTD, but has yet to be explored as potential biomarker. METHODS Plasma GFAP and neurofilament light chain (NfL) concentration were measured in 469 individuals enrolled in the Genetic FTD Initiative: 114 C9orf72 expansion carriers (74 presymptomatic, 40 symptomatic), 119 GRN mutation carriers (88 presymptomatic, 31 symptomatic), 53 MAPT mutation carriers (34 presymptomatic, 19 symptomatic) and 183 non-carrier controls. Biomarker measures were compared between groups using linear regression models adjusted for age and sex with family membership included as random effect. Participants underwent standardised clinical assessments including the Mini-Mental State Examination (MMSE), Frontotemporal Lobar Degeneration-Clinical Dementia Rating scale and MRI. Spearman's correlation coefficient was used to investigate the relationship of plasma GFAP to clinical and imaging measures. RESULTS Plasma GFAP concentration was significantly increased in symptomatic GRN mutation carriers (adjusted mean difference from controls 192.3 pg/mL, 95% CI 126.5 to 445.6), but not in those with C9orf72 expansions (9.0, -61.3 to 54.6), MAPT mutations (12.7, -33.3 to 90.4) or the presymptomatic groups. GFAP concentration was significantly positively correlated with age in both controls and the majority of the disease groups, as well as with NfL concentration. In the presymptomatic period, higher GFAP concentrations were correlated with a lower cognitive score (MMSE) and lower brain volume, while in the symptomatic period, higher concentrations were associated with faster rates of atrophy in the temporal lobe. CONCLUSIONS Raised GFAP concentrations appear to be unique to GRN-related FTD, with levels potentially increasing just prior to symptom onset, suggesting that GFAP may be an important marker of proximity to onset, and helpful for forthcoming therapeutic prevention trials.
Collapse
Affiliation(s)
- Carolin Heller
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Martha S Foiani
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Katrina Moore
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian Convery
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Mollie Neason
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK.,Centre for Medical Image Computing, University College London, London, UK
| | - David Thomas
- Neuradiological Academic Unit, UCL Queen Square Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Ione Oc Woollacott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Rachelle Shafei
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - John C Van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, País Vasco, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire du CHU de Québec, Département des Sciences Neurologiques, Université Laval, Québec, Québec, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital-Huddinge, Stockholm, Sweden
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan, Italy.,Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Fabrizio Tagliavini
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Simon Ducharme
- Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | | | - Alex Gerhard
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Amanda J Heslegrave
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | | |
Collapse
|
74
|
Diagnostic-prognostic value and electrophysiological correlates of CSF biomarkers of neurodegeneration and neuroinflammation in amyotrophic lateral sclerosis. J Neurol 2020; 267:1699-1708. [DOI: 10.1007/s00415-020-09761-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
|
75
|
de Majo M, Koontz M, Rowitch D, Ullian EM. An update on human astrocytes and their role in development and disease. Glia 2020; 68:685-704. [PMID: 31926040 DOI: 10.1002/glia.23771] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/25/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
Human astrocytes provide trophic as well as structural support to the surrounding brain cells. Furthermore, they have been implicated in many physiological processes important for central nervous system function. Traditionally astrocytes have been considered to be a homogeneous class of cells, however, it has increasingly become more evident that astrocytes can have very different characteristics in different regions of the brain, or even within the same region. In this review we will discuss the features of human astrocytes, their heterogeneity, and their generation during neurodevelopment and the extraordinary progress that has been made to model these fascinating cells in vitro, mainly from induced pluripotent stem cells. Astrocytes' role in disease will also be discussed with a particular focus on their role in neurodegenerative disorders. As outlined here, astrocytes are important for the homeostasis of the central nervous system and understanding their regional specificity is a priority to elucidate the complexity of the human brain.
Collapse
Affiliation(s)
- Martina de Majo
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| | - Mark Koontz
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| | - David Rowitch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.,Department of Pediatrics, University of California, San Francisco, San Francisco, California.,Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
76
|
Congenic expression of poly-GA but not poly-PR in mice triggers selective neuron loss and interferon responses found in C9orf72 ALS. Acta Neuropathol 2020; 140:121-142. [PMID: 32562018 PMCID: PMC7360660 DOI: 10.1007/s00401-020-02176-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
Expansion of a (G4C2)n repeat in C9orf72 causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), but the link of the five repeat-encoded dipeptide repeat (DPR) proteins to neuroinflammation, TDP-43 pathology, and neurodegeneration is unclear. Poly-PR is most toxic in vitro, but poly-GA is far more abundant in patients. To directly compare these in vivo, we created congenic poly-GA and poly-PR mice. 40% of poly-PR mice were affected with ataxia and seizures, requiring euthanasia by 6 weeks of age. The remaining poly-PR mice were asymptomatic at 14 months of age, likely due to an 80% reduction of the transgene mRNA in this subgroup. In contrast, all poly-GA mice showed selective neuron loss, inflammation, as well as muscle denervation and wasting requiring euthanasia before 7 weeks of age. In-depth analysis of peripheral organs and blood samples suggests that peripheral organ failure does not drive these phenotypes. Although transgene mRNA levels were similar between poly-GA and affected poly-PR mice, poly-GA aggregated far more abundantly than poly-PR in the CNS and was also found in skeletal muscle. In addition, TDP-43 and other disease-linked RNA-binding proteins co-aggregated in rare nuclear inclusions in the hippocampus and frontal cortex only in poly-GA mice. Transcriptome analysis revealed activation of an interferon-responsive pro-inflammatory microglial signature in end-stage poly-GA but not poly-PR mice. This signature was also found in all ALS patients and enriched in C9orf72 cases. In summary, our rigorous comparison of poly-GA and poly-PR toxicity in vivo indicates that poly-GA, but not poly-PR at the same mRNA expression level, promotes interferon responses in C9orf72 disease and contributes to TDP-43 abnormalities and neuron loss selectively in disease-relevant regions.
Collapse
|
77
|
Oeckl P, Weydt P, Thal DR, Weishaupt JH, Ludolph AC, Otto M. Proteomics in cerebrospinal fluid and spinal cord suggests UCHL1, MAP2 and GPNMB as biomarkers and underpins importance of transcriptional pathways in amyotrophic lateral sclerosis. Acta Neuropathol 2020; 139:119-134. [PMID: 31701227 DOI: 10.1007/s00401-019-02093-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Accepted: 11/01/2019] [Indexed: 01/09/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease and the proteins and pathways involved in the pathophysiology are not fully understood. Even less is known about the preclinical disease phase. To uncover new ALS-related proteins and pathways, we performed a comparative proteomic analysis in cerebrospinal fluid (CSF) of asymptomatic (n = 14) and symptomatic (n = 14) ALS mutation carriers and sporadic ALS patients (n = 12) as well as post-mortem human spinal cord tissue (controls: n = 7, ALS, n = 8). Using a CSF-optimized proteomic workflow, we identified novel (e.g., UCHL1, MAP2, CAPG, GPNMB, HIST1H4A, HIST1H2B) and well-described (e.g., NEFL, NEFH, NEFM, CHIT1, CHI3L1) protein level changes in CSF of sporadic and genetic ALS patients with enrichment of proteins related to transcription, cell cycle and lipoprotein remodeling (total protein IDs: 2303). No significant alteration was observed in asymptomatic ALS mutation carriers representing the prodromal disease phase. We confirmed UCHL1, MAP2, CAPG and GPNMB as novel biomarker candidates for ALS in an independent validation cohort of patients (n = 117) using multiple reaction monitoring. In spinal cord tissue, 292 out of 6810 identified proteins were significantly changed in ALS with enrichment of proteins involved in mRNA splicing and of the neurofilament compartment. In conclusion, our proteomic data in asymptomatic ALS mutation carriers support the hypothesis of a sudden disease onset instead of a long preclinical phase. Both CSF and tissue proteomic data indicate transcriptional pathways to be amongst the most affected. UCHL1, MAP2 and GPNMB are promising ALS biomarker candidates which might provide additional value to the established neurofilaments in patient follow-up and clinical trials.
Collapse
Affiliation(s)
- Patrick Oeckl
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Patrick Weydt
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081, Ulm, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Dietmar R Thal
- Laboratory of Neuropathology, Institute of Pathology, Ulm University, Ulm, Germany
- Department of Imaging and Pathology, KU Leuven and Department of Pathology, UZ Leuven, Louvain, Belgium
| | - Jochen H Weishaupt
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Albert C Ludolph
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Oberer Eselsberg 45, 89081, Ulm, Germany.
| |
Collapse
|
78
|
Abu-Rumeileh S, Steinacker P, Polischi B, Mammana A, Bartoletti-Stella A, Oeckl P, Baiardi S, Zenesini C, Huss A, Cortelli P, Capellari S, Otto M, Parchi P. CSF biomarkers of neuroinflammation in distinct forms and subtypes of neurodegenerative dementia. Alzheimers Res Ther 2019; 12:2. [PMID: 31892365 PMCID: PMC6937795 DOI: 10.1186/s13195-019-0562-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND In neurodegenerative dementias (NDs) such as prion disease, Alzheimer's disease (AD), and frontotemporal lobar degeneration (FTLD), protein misfolding leads to the tissue deposition of protein aggregates which, in turn, trigger neuroinflammation and neurodegeneration. Cerebrospinal fluid (CSF) biomarkers have the potential to reflect different aspects of these phenomena across distinct clinicopathological subtypes and disease stages. METHODS We investigated CSF glial markers, namely chitotriosidase 1 (CHIT1), chitinase-3-like protein 1 (YKL-40) and glial fibrillary acidic protein (GFAP) in prion disease subtypes (n = 101), AD (n = 40), clinicopathological subgroups of FTLD (n = 72), and controls (n = 40) using validated, commercially available ELISA assays. We explored glial biomarker levels' associations with disease variables and neurodegenerative CSF biomarkers and evaluated their diagnostic accuracy. The genotype of the CHIT1 rs3831317 polymorphic site was also analyzed. RESULTS Each ND group showed increased levels of CHIT1, YKL-40, and GFAP compared to controls with a difference between prion disease and AD or FTLD limited to YKL-40, which showed higher values in the former group. CHIT1 levels were reduced in both heterozygotes and homozygotes for the CHIT1 24-bp duplication (rs3831317) in FTLD and controls, but this effect was less significant in AD and prion disease. After stratification according to molecular subgroups, we demonstrated (i) an upregulation of all glial markers in Creutzfeldt-Jakob disease VV2 compared to other disease subtypes, (ii) a difference in CHIT1 levels between FTLD with TAU and TDP43 pathology, and (iii) a marked increase of YKL-40 in FTLD with amyotrophic lateral sclerosis (ALS) in comparison with FTLD without ALS. In prion disease, glial markers correlated with disease stage and were already elevated in one pre-symptomatic case of Gerstmann-Sträussler-Scheinker disease. Regarding the diagnostic value, YKL-40 was the only glial marker that showed a moderate accuracy in the distinction between controls and NDs. CONCLUSIONS NDs share a CSF profile characterized by increased levels of CSF CHIT1, YKL-40, and GFAP, which likely reflects a common neuroinflammatory response to protein misfolding and aggregation. CSF glial markers of neuroinflammation demonstrate limited diagnostic value but have some potential for monitoring the clinical and, possibly, preclinical phases of NDs.
Collapse
Affiliation(s)
- Samir Abu-Rumeileh
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy
| | - Petra Steinacker
- Department of Neurology, Ulm University Hospital, 89073 Ulm, Germany
| | - Barbara Polischi
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Angela Mammana
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Anna Bartoletti-Stella
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Patrick Oeckl
- Department of Neurology, Ulm University Hospital, 89073 Ulm, Germany
| | - Simone Baiardi
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy
| | - Corrado Zenesini
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - André Huss
- Department of Neurology, Ulm University Hospital, 89073 Ulm, Germany
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Sabina Capellari
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, 89073 Ulm, Germany
| | - Piero Parchi
- Ospedale Bellaria, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
79
|
Morelli C, Tiloca C, Colombrita C, Zambon A, Soranna D, Lafronza A, Solca F, Carelli L, Poletti B, Doretti A, Verde F, Maderna L, Ticozzi N, Ratti A, Silani V. CSF angiogenin levels in amyotrophic lateral Sclerosis-Frontotemporal dementia spectrum. Amyotroph Lateral Scler Frontotemporal Degener 2019; 21:63-69. [DOI: 10.1080/21678421.2019.1704016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- C. Morelli
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - C. Tiloca
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - C. Colombrita
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - A. Zambon
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy,
| | - D. Soranna
- Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - A. Lafronza
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - F. Solca
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - L. Carelli
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - B. Poletti
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - A. Doretti
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - F. Verde
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - L. Maderna
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
| | - N. Ticozzi
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy,
| | - A. Ratti
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy,
| | - V. Silani
- Department of Neurology-Stroke Unit and Laboratory of Neurosciences, Istituto Auxologico Italiano, IRCCS, Milan, Italy,
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy,
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
80
|
Thompson AG, Gray E, Bampton A, Raciborska D, Talbot K, Turner MR. CSF chitinase proteins in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2019; 90:1215-1220. [PMID: 31123140 DOI: 10.1136/jnnp-2019-320442] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the classifier performance, clinical and biochemical correlations of cerebrospinal fluid (CSF) levels of the chitinase proteins Chitotriosidase-1 (CHIT1), Chitinase-3-like protein 1 (CHI3L1) and Chitinase-3-like protein 2 (CHI3L2) in amyotrophic lateral sclerosis (ALS). METHODS CSF levels of CHIT1, CHI3L1, CHI3L2, phosphorylated neurofilament heavy chain (pNFH) and C-reactive protein were measured by ELISA in a longitudinal cohort of patients with ALS (n=82), primary lateral sclerosis (PLS, n=10), ALS-mimic conditions (n=12), healthy controls (n=25) and asymptomatic carriers of ALS-causing genetic mutations (AGC; n=5). RESULTS CSF CHIT1, CHI3L1 and CHI3L2 were elevated in patients with ALS compared with healthy controls (p<0.001) and ALS-mimics (CHIT1, p<0.001; CHI3L1, p=0.017; CHI3L2, p<0.001). CHIT1 and CHI3L2 were elevated in ALS compared with PLS (CHIT1, p=0.021; CHI3L1, p=0.417; CHI3L2, p<0.001). Chitinase levels were similar in AGCs and healthy controls. Chitinase proteins distinguished ALS from healthy controls (area under the curve (AUC): CHIT1 0.92; CHI3L1 0.80; CHI3L2 0.90), mimics (AUC: CHIT1 0.84; CHI3L1 0.73; CHI3L2 0.88) and, to a lesser extent, PLS (AUC: CHIT 0.73; CHI3L1 0.51; CHI3L2 0.82) but did not outperform pNFH. CHIT1 and CHI3L2 correlated with disease progression rate (Pearson's r=0.49, p<0.001; r=0.42, p<0.001, respectively). CHI3L1 correlated with degree of cognitive dysfunction (r=-0.25, p=0.038). All chitinases correlated with pNFH. CHIT1 levels were associated with survival in multivariate models. Chitinase levels were longitudinally stable. CONCLUSIONS CSF chitinase proteins may have limited value as independent diagnostic and stratification biomarkers in ALS, but offer a window into non-autonomous mechanisms of motor neuronal loss in ALS, specifically in assessing response to therapies targeting neuroinflammatory pathways.
Collapse
Affiliation(s)
| | - Elizabeth Gray
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Alexander Bampton
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | | | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| |
Collapse
|
81
|
Fraga VG, Magalhães CA, Loures CDMG, de Souza LC, Guimarães HC, Zauli DAG, Carvalho MDG, Ferreira CN, Caramelli P, de Sousa LP, Gomes KB. Inflammatory and Pro-resolving Mediators in Frontotemporal Dementia and Alzheimer's Disease. Neuroscience 2019; 421:123-135. [PMID: 31654714 DOI: 10.1016/j.neuroscience.2019.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 01/06/2023]
Abstract
Chronic inflammation contributes to neuronal death in Alzheimer's disease (AD) and frontotemporal dementia (FTD). Here we evaluated inflammatory and pro-resolving mediators in AD and behavioural variant of FTD (bvFTD) patients compared with controls, since neuroinflamamtion is a common feature in both diseases. Ninety-eight subjects were included in this study, divided into AD (n = 32), bvFTD (n = 30), and control (n = 36) groups. The levels of hsCRP, IL-1β, IL-6, TNF, and TGF-β1, as well as annexin A1 (AnxA1) and lipoxin A4 (LXA4) were measured in blood and cerebrospinal fluid (CSF). The expression profile of AnxA1 was evaluated in peripheral blood mononuclear cells (PBMCs) as well the distribution of ANXA1 rs2611228 polymorphism. We found reduced peripheral levels of hsCRP and TNF in AD compared with bvFTD patients and controls, and increased levels of TGF-β1 in AD compared to controls. Moreover, reduced plasma levels of AnxA1 were observed in bvFTD compared to AD and controls. There was a significant cleavage of AnxA1 in PBMCs in both dementia groups. The results suggest differential regulation of inflammatory and pro-resolving mediators in bvFTD and AD, while AnxA1 cleavage may impair pro-resolving mechanisms in both groups.
Collapse
Affiliation(s)
- Vanêssa Gomes Fraga
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carolina Antunes Magalhães
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina de Mello Gomide Loures
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Cruz de Souza
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Henrique Cerqueira Guimarães
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paulo Caramelli
- Departamento de Clínca Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lirlândia Pires de Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
82
|
|
83
|
|
84
|
Halpern M, Brennand KJ, Gregory J. Examining the relationship between astrocyte dysfunction and neurodegeneration in ALS using hiPSCs. Neurobiol Dis 2019; 132:104562. [PMID: 31381978 DOI: 10.1016/j.nbd.2019.104562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and fatal neurodegenerative disease for which the causes of disease onset and progression remain unclear. Recent advances in human induced pluripotent stem cell (hiPSC)-based models permit the study of the genetic factors associated with ALS in patient-derived neural cell types, including motor neurons and glia. While astrocyte dysfunction has traditionally been thought to exacerbate disease progression, astrocytic dysfunction may play a more direct role in disease initiation and progression. Such non-cell autonomous mechanisms expand the potential targets of therapeutic intervention, but only a handful of ALS risk-associated genes have been examined for their impact on astrocyte dysfunction and neurodegeneration. This review summarizes what is currently known about astrocyte function in ALS and suggests ways in which hiPSC-based models can be used to more effectively study the role of astrocytes in neurodegenerative disease.
Collapse
Affiliation(s)
- Madeline Halpern
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| | - James Gregory
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, United States of America.
| |
Collapse
|
85
|
Häussler RS, Bendes A, Iglesias M, Sanchez-Rivera L, Dodig-Crnković T, Byström S, Fredolini C, Birgersson E, Dale M, Edfors F, Fagerberg L, Rockberg J, Tegel H, Uhlén M, Qundos U, Schwenk JM. Systematic Development of Sandwich Immunoassays for the Plasma Secretome. Proteomics 2019; 19:e1900008. [PMID: 31278833 DOI: 10.1002/pmic.201900008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The plasma proteome offers a clinically useful window into human health. Recent advances from highly multiplexed assays now call for appropriate pipelines to validate individual candidates. Here, a workflow is developed to build dual binder sandwich immunoassays (SIA) and for proteins predicted to be secreted into plasma. Utilizing suspension bead arrays, ≈1800 unique antibody pairs are first screened against 209 proteins with recombinant proteins as well as EDTA plasma. Employing 624 unique antibodies, dilution-dependent curves in plasma and concentration-dependent curves of full-length proteins for 102 (49%) of the targets are obtained. For 22 protein assays, the longitudinal, interindividual, and technical performance is determined in a set of plasma samples collected from 18 healthy subjects every third month over 1 year. Finally, 14 of these assays are compared with with SIAs composed of other binders, proximity extension assays, and affinity-free targeted mass spectrometry. The workflow provides a multiplexed approach to screen for SIA pairs that suggests using at least three antibodies per target. This design is applicable for a wider range of targets of the plasma proteome, and the assays can be applied for discovery but also to validate emerging candidates derived from other platforms.
Collapse
Affiliation(s)
- Ragna S Häussler
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Annika Bendes
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - MariaJesus Iglesias
- Division of Cellular and Clinical Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
- K.G. Jebsen - Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, 9010, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, 9010, Tromsø, Norway
| | - Laura Sanchez-Rivera
- Division of Cellular and Clinical Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Tea Dodig-Crnković
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Sanna Byström
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Claudia Fredolini
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Elin Birgersson
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Matilda Dale
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Fredrik Edfors
- Division of Systems Biology, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Linn Fagerberg
- Division of Systems Biology, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| | - Johan Rockberg
- Division of Protein Technology, Department of Protein Science, KTH - Royal Institute of Technology, 106 91, Stockholm, Sweden
| | - Hanna Tegel
- Division of Protein Technology, Department of Protein Science, KTH - Royal Institute of Technology, 106 91, Stockholm, Sweden
| | - Mathias Uhlén
- Division of Systems Biology, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2970, Hørsholm, Denmark
| | | | - Jochen M Schwenk
- Division of Affinity Proteomics, Science for Life Laboratory, KTH - Royal Institute of Technology, Box 1031, 171 21, Solna, Sweden
| |
Collapse
|
86
|
Rué L, Oeckl P, Timmers M, Lenaerts A, van der Vos J, Smolders S, Poppe L, de Boer A, Van Den Bosch L, Van Damme P, Weishaupt JH, Ludolph AC, Otto M, Robberecht W, Lemmens R. Reduction of ephrin-A5 aggravates disease progression in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2019; 7:114. [PMID: 31300041 PMCID: PMC6626434 DOI: 10.1186/s40478-019-0759-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/22/2019] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons in the brainstem, spinal cord and motor cortex. ALS is characterized by genetic and clinical heterogeneity, suggesting the existence of genetic factors that modify the phenotypic expression of the disease. We previously identified the axonal guidance EphA4 receptor, member of the Eph-ephrin system, as an ALS disease-modifying factor. EphA4 genetic inhibition rescued the motor neuron phenotype in zebrafish and a rodent model of ALS. Preventing ligands from binding to the EphA4 receptor also successfully improved disease, suggesting a role for EphA4 ligands in ALS. One particular ligand, ephrin-A5, is upregulated in reactive astrocytes after acute neuronal injury and inhibits axonal regeneration. Moreover, it plays a role during development in the correct pathfinding of motor axons towards their target limb muscles. We hypothesized that a constitutive reduction of ephrin-A5 signalling would benefit disease progression in a rodent model for ALS. We discovered that in the spinal cord of control and symptomatic ALS mice ephrin-A5 was predominantly expressed in neurons. Surprisingly, reduction of ephrin-A5 levels in SOD1G93A mice accelerated disease progression and reduced survival without affecting disease onset, motor neuron numbers or innervated neuromuscular junctions in symptomatic mice. These findings suggest ephrin-A5 as a modifier of disease progression that might play a role in the later stages of the disease. Similarly, we identified a more aggressive disease progression in patients with lower ephrin-A5 protein levels in the cerebrospinal fluid without modifying disease onset. In summary, we identified reduced expression of ephrin-A5 to accelerate disease progression in a mouse model of ALS as well as in humans. Combined with our previous findings on the role of EphA4 in ALS our current data suggests different contribution for various members of the Eph-ephrin system in the pathophysiology of a motor neuron disease.
Collapse
|
87
|
Villar-Piqué A, Schmitz M, Hermann P, Goebel S, Bunck T, Varges D, Ferrer I, Riggert J, Llorens F, Zerr I. Plasma YKL-40 in the spectrum of neurodegenerative dementia. J Neuroinflammation 2019; 16:145. [PMID: 31299989 PMCID: PMC6624942 DOI: 10.1186/s12974-019-1531-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Background Increased plasma YKL-40 has been reported in Alzheimer’s disease (AD), but its levels in other neurodegenerative diseases are unknown. Here, we aimed to investigate plasma YKL-40 in the spectrum of neurodegenerative dementias. Methods YKL-40 was quantified in the plasma of 315 cases, including healthy controls (HC), neurological disease controls (ND), AD, vascular dementia (VaD), frontotemporal dementia (FTD), sporadic Creutzfeldt-Jakob disease (CJD) and Lewy body dementia (LBD). Diagnostic accuracy in the differential diagnostic context and influence of age and gender was assessed. Results Highest YKL-40 levels were detected in CJD, followed by LBD, VaD, AD, FTD, ND and HC. YKL-40 was associated to age but not to sex. After controlling for age, YKL-40 was significantly elevated in CJD compared to HC (p < 0.001), ND, AD and VaD (p < 0.01) and in LBD compared to HC (p < 0.05). In CJD, YKL-40 concentrations were significantly higher at late disease stages. Conclusions Plasma YKL-40 is significantly elevated in CJD regardless of clinical and genetic parameters, with moderate diagnostic accuracy in the discrimination from control cases. Our study discards a potential use of this biomarker in the differential diagnostic context but opens the possibility to be explored as a marker for CJD monitoring.
Collapse
Affiliation(s)
- Anna Villar-Piqué
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany.
| | - Matthias Schmitz
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Peter Hermann
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany
| | - Stefan Goebel
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany
| | - Timothy Bunck
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany
| | - Daniela Varges
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany
| | - Isidre Ferrer
- Network Center for Biomedical Research in Neurodegenerative Diseases, (CIBERNED), Institute Carlos III, Ministry of Health, Feixa Llarga s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain
| | - Joachim Riggert
- Department of Transfusion Medicine, University Medical School, Göttingen, Germany
| | - Franc Llorens
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany. .,Network Center for Biomedical Research in Neurodegenerative Diseases, (CIBERNED), Institute Carlos III, Ministry of Health, Feixa Llarga s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Spain. .,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Robert Koch 40, 37075, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
88
|
Baldacci F, Lista S, Palermo G, Giorgi FS, Vergallo A, Hampel H. The neuroinflammatory biomarker YKL-40 for neurodegenerative diseases: advances in development. Expert Rev Proteomics 2019; 16:593-600. [PMID: 31195846 DOI: 10.1080/14789450.2019.1628643] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Neuroinflammation is a common pathophysiological mechanism in neurodegenerative diseases (ND). Cerebrospinal fluid (CSF) YKL-40 has recently been candidated as a neuroinflammatory biomarker of ND. Areas covered: We provide an update on the role of CSF YKL-40 as a pathophysiological biomarker of ND. YKL-40 may discriminate Alzheimer's disease (AD) from controls and may predict the progression from the early preclinical to the late dementia stage. In genetic AD, YKL-40 increases decades before the clinical onset. It does not seem a specific biomarker of a certain ND although sporadic Creutzfeldt-Jacob disease shows the highest YKL-40 concentrations. YKL-40 may discriminate between amyotrophic lateral sclerosis (ALS) and ALS-mimics. YKL-40 is potentially associated with the rate of ALS progression. YKL-40 correlates with biomarkers of neuronal injury, large axonal damage and synaptic disruption in various ND. It is not associated with the presence of the APOE-ε4 allele whereas possibly linked to aging, female sex, Hispanic ethnicity and some genetic variants of the chitinase-3-like 1 locus. Expert opinion: There is growing evidence expanding the relevance of CSF YKL-40 as a pathophysiological biomarker for ND. Patients showing high YKL-40 levels might benefit from targeted clinical trials that use compounds acting against neuroinflammatory mechanisms, independently of the initial clinical diagnosis of ND.
Collapse
Affiliation(s)
- Filippo Baldacci
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy.,b Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital , F-75013, Paris , France.,c Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital , F-75013, Paris , France.,d Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP , Boulevard de l'hôpital , F-75013, Paris , France
| | - Simone Lista
- b Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital , F-75013, Paris , France.,c Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital , F-75013, Paris , France.,d Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP , Boulevard de l'hôpital , F-75013, Paris , France
| | - Giovanni Palermo
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Filippo Sean Giorgi
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Andrea Vergallo
- b Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital , F-75013, Paris , France.,c Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital , F-75013, Paris , France.,d Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP , Boulevard de l'hôpital , F-75013, Paris , France
| | - Harald Hampel
- b Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital , F-75013, Paris , France
| |
Collapse
|
89
|
McCombe PA, Henderson RD, Lee A, Lee JD, Woodruff TM, Restuadi R, McRae A, Wray NR, Ngo S, Steyn FJ. Gut microbiota in ALS: possible role in pathogenesis? Expert Rev Neurother 2019; 19:785-805. [PMID: 31122082 DOI: 10.1080/14737175.2019.1623026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: The gut microbiota has important roles in maintaining human health. The microbiota and its metabolic byproducts could play a role in the pathogenesis of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Areas covered: The authors evaluate the methods of assessing the gut microbiota, and also review how the gut microbiota affects the various physiological functions of the gut. The authors then consider how gut dysbiosis could theoretically affect the pathogenesis of ALS. They present the current evidence regarding the composition of the gut microbiota in ALS and in rodent models of ALS. Finally, the authors review therapies that could improve gut dysbiosis in the context of ALS. Expert opinion: Currently reported studies suggest some instances of gut dysbiosis in ALS patients and mouse models; however, these studies are limited, and more information with well-controlled larger datasets is required to make a definitive judgment about the role of the gut microbiota in ALS pathogenesis. Overall this is an emerging field that is worthy of further investigation. The authors advocate for larger studies using modern metagenomic techniques to address the current knowledge gaps.
Collapse
Affiliation(s)
- Pamela A McCombe
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,School of Medicine, The University of Queensland , Brisbane , Australia
| | - Robert D Henderson
- Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,School of Medicine, The University of Queensland , Brisbane , Australia.,Queensland Brain Institute, The University of Queensland , Brisbane , Australia
| | - Aven Lee
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Restuadi Restuadi
- Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Allan McRae
- Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Naomi R Wray
- Queensland Brain Institute, The University of Queensland , Brisbane , Australia.,Institute for Molecular Bioscience, The University of Queensland , Brisbane , Australia
| | - Shyuan Ngo
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,Queensland Brain Institute, The University of Queensland , Brisbane , Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane , Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland , Brisbane , Australia.,Wesley Medical Research, Level 8 East Wing, The Wesley Hospital , Brisbane , Australia.,Department of Neurology, Royal Brisbane & Women's Hospital , Brisbane , Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane , Australia
| |
Collapse
|
90
|
An update on genetic frontotemporal dementia. J Neurol 2019; 266:2075-2086. [PMID: 31119452 PMCID: PMC6647117 DOI: 10.1007/s00415-019-09363-4] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia (FTD) is a highly heritable group of neurodegenerative disorders, with around 30% of patients having a strong family history. The majority of that heritability is accounted for by autosomal dominant mutations in the chromosome 9 open reading frame 72 (C9orf72), progranulin (GRN), and microtubule-associated protein tau (MAPT) genes, with mutations more rarely seen in a number of other genes. This review will discuss the recent updates in the field of genetic FTD. Age at symptom onset in genetic FTD is variable with recently identified genetic modifiers including TMEM106B (in GRN carriers particularly) and a polymorphism at a locus containing two overlapping genes LOC101929163 and C6orf10 (in C9orf72 carriers). Behavioural variant FTD (bvFTD) is the most common diagnosis in each of the genetic groups, although in C9orf72 carriers amyotrophic lateral sclerosis either alone, or with bvFTD, is also common. An atypical neuropsychiatric presentation is also seen in C9orf72 carriers and family members of carriers are at greater risk of psychiatric disorders including schizophrenia and autistic spectrum disorders. Large natural history studies of presymptomatic genetic FTD are now underway both in Europe/Canada (GENFI—the Genetic FTD Initiative) and in the US (ARTFL/LEFFTDS study), collaborating together under the banner of the FTD Prevention Initiative (FPI). These studies are taking forward the validation of cognitive, imaging and fluid biomarkers that aim to robustly measure disease onset, staging and progression in genetic FTD. Grey matter changes on MRI and hypometabolism on FDG-PET are seen at least 10 years before symptom onset with white matter abnormalities seen earlier, but the pattern and exact timing of changes differ between different genetic groups. In contrast, tau PET has yet to show promise in genetic FTD. Three key fluid biomarkers have been identified so far that are likely to be helpful in clinical trials—CSF or blood neurofilament light chain levels (in all groups), CSF or blood progranulin levels (in GRN carriers) and CSF poly(GP) dipeptide repeat protein levels (in C9orf72 carriers). Increased knowledge about genetic FTD has led to more clinical presymptomatic genetic testing but this has not yet been mirrored in the development of either an accepted FTD-specific testing protocol or provision of appropriate psychological support mechanisms for those living through the at-risk phase. This will become even more relevant as disease-modifying therapy trials start in each of the genetic groups over the next few years.
Collapse
|
91
|
Rostalski H, Leskelä S, Huber N, Katisko K, Cajanus A, Solje E, Marttinen M, Natunen T, Remes AM, Hiltunen M, Haapasalo A. Astrocytes and Microglia as Potential Contributors to the Pathogenesis of C9orf72 Repeat Expansion-Associated FTLD and ALS. Front Neurosci 2019; 13:486. [PMID: 31156371 PMCID: PMC6529740 DOI: 10.3389/fnins.2019.00486] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) are neurodegenerative diseases with a complex, but often overlapping, genetic and pathobiological background and thus they are considered to form a disease spectrum. Although neurons are the principal cells affected in FTLD and ALS, increasing amount of evidence has recently proposed that other central nervous system-resident cells, including microglia and astrocytes, may also play roles in neurodegeneration in these diseases. Therefore, deciphering the mechanisms underlying the disease pathogenesis in different types of brain cells is fundamental in order to understand the etiology of these disorders. The major genetic cause of FTLD and ALS is a hexanucleotide repeat expansion (HRE) in the intronic region of the C9orf72 gene. In neurons, specific pathological hallmarks, including decreased expression of the C9orf72 RNA and proteins and generation of toxic RNA and protein species, and their downstream effects have been linked to C9orf72 HRE-associated FTLD and ALS. In contrast, it is still poorly known to which extent these pathological changes are presented in other brain cells. Here, we summarize the current literature on the potential role of astrocytes and microglia in C9orf72 HRE-linked FTLD and ALS and discuss their possible phenotypic alterations and neurotoxic mechanisms that may contribute to neurodegeneration in these diseases.
Collapse
Affiliation(s)
- Hannah Rostalski
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stina Leskelä
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nadine Huber
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kasper Katisko
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Antti Cajanus
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Anne M Remes
- Medical Research Center, Oulu University Hospital, Oulu, Finland.,Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
92
|
Borroni B, Alberici A, Buratti E. Review: Molecular pathology of frontotemporal lobar degenerations. Neuropathol Appl Neurobiol 2019; 45:41-57. [DOI: 10.1111/nan.12534] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023]
Affiliation(s)
- B. Borroni
- Neurology Clinic; Department of Clinical and Experimental Sciences; University of Brescia; Brescia Italy
| | - A. Alberici
- Neurology Clinic; Department of Clinical and Experimental Sciences; University of Brescia; Brescia Italy
| | - E. Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB); Trieste Italy
| |
Collapse
|
93
|
Vucic S. Differences in inflammatory profiles between ALS and FTD. J Neurol Neurosurg Psychiatry 2019; 90:1. [PMID: 30224547 DOI: 10.1136/jnnp-2018-319377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Steve Vucic
- Western Clinical School, University of Sydney, Sydney, NSW 2145, Australia
| |
Collapse
|