1
|
Carrazana E, Montalbán-Gutiérrez L, Chana-Cuevas P, Salvadores N. Advancing Parkinson's diagnosis: seed amplification assay for α-synuclein detection in minimally invasive samples. Mol Cell Biochem 2025:10.1007/s11010-024-05190-y. [PMID: 39760833 DOI: 10.1007/s11010-024-05190-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by tremor, rigidity, and bradykinesia, beginning with early loss of dopaminergic neurons in the ventrolateral substantia nigra and advancing to broader neurodegeneration in the midbrain. The clinical heterogeneity of PD and the lack of specific diagnostic tests present significant challenges, highlighting the need for reliable biomarkers for early diagnosis. Alpha-synuclein (α-Syn), a protein aggregating into Lewy bodies and neurites in PD patients, has emerged as a key biomarker due to its central role in PD pathophysiology and potential to reflect pathological processes. Additionally, α-Syn allows earlier differentiation between PD and other neurodegenerative disorders with similar symptoms. Currently, detection of α-Syn pathology in post-mortem brain tissue remains the primary means of achieving a conclusive diagnosis, often revealing significant misdiagnoses. Seed amplification assay (SAA), initially developed for prion diseases, has been adapted to detect α-Syn aggregates in cerebrospinal fluid, showing promise for early diagnosis. Recent studies have demonstrated that SAA can also detect α-Syn aggregates in peripheral samples collected via minimally invasive procedures, such as skin, olfactory mucosa, saliva, and blood. However, the lack of standardized protocols limits clinical application. Standardizing protocols is essential to improve assay reliability and enable accurate patient identification for emerging therapies. This review examines studies on SAA for detecting α-Syn aggregates in minimally invasive samples, focusing on sample collection, processing, and reaction conditions.
Collapse
Affiliation(s)
- Elizabeth Carrazana
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile
| | - Leonardo Montalbán-Gutiérrez
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile
- Escuela de Medicina, Facultad de Medicina y Ciencias de La Salud, Universidad Mayor, Temuco, Chile
| | - Pedro Chana-Cuevas
- Centro de Trastornos del Movimiento (CETRAM), Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Natalia Salvadores
- Neurodegenerative Diseases Laboratory, Center for Biomedicine, Universidad Mayor, Avenida Alemania 0281, 4780000, Temuco, La Araucanía, Chile.
- Escuela de Medicina, Facultad de Medicina y Ciencias de La Salud, Universidad Mayor, Temuco, Chile.
| |
Collapse
|
2
|
Salciccia C, Costanzo M, Ruocco G, Porreca F, Vivacqua G, Fabbrini G, Belvisi D, Ladogana A, Poleggi A. Proteopathic seed amplification assays in easily accessible specimens for human synucleinopathies, tauopathies and prionopathies: a Scoping Review. Neurosci Biobehav Rev 2024:105997. [PMID: 39746590 DOI: 10.1016/j.neubiorev.2024.105997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/30/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
A hallmark event in neurodegenerative diseases is represented by the misfolding, aggregation and accumulation of proteins, leading to cellular and network dysfunction preceding the development of clinical symptoms by years. Early diagnosis represents a crucial issue in the field of neuroscience as it offers the potential to utilize this therapeutic window in the future to manage disease-modifying therapy. Seed amplification assays, including Real-Time Quaking-Induced Conversion (RT-QuIC) and Protein Misfolding Cyclic Amplification (PMCA), have emerged in recent years as innovative techniques developed to detect minute amounts of amyloidogenic proteins. These techniques can utilize various biological fluids and tissues, with most evidence to date regarding their potential diagnostic use focusing on cerebrospinal fluid. In this scoping review, we aimed to investigate and discuss the available evidence regarding the diagnostic use of these assays on easily accessible biological fluids and tissues in patients affected by synucleinopathies, tauopathies or prion diseases. From a systematic search on two databases, Scopus and Pubmed, we identified 45 studies. Although most identified studies have used skin and olfactory mucosa as biological samples, there is preliminary evidence suggesting the potential implementation of these techniques using fluids as blood, saliva and tears. The results achieved so far, as well as methodological aspects and limitations to overcome, are discussed.
Collapse
Affiliation(s)
- Clara Salciccia
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Matteo Costanzo
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy; Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, 00185, Rome (RM), Italy
| | - Giulia Ruocco
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, 00185, Rome (RM), Italy
| | - Flavia Porreca
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Giorgio Vivacqua
- Department of Microscopic and Ultrastructural Anatomy-Integrated Research Center (PRAAB), Campus Biomedico University of Rome, 00128 Rome, Italy
| | - Giovanni Fabbrini
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, 00185, Rome (RM), Italy; IRCCS Neuromed, Via Atinense 18, 86077, Pozzilli (IS), Italy
| | - Daniele Belvisi
- Department of Human Neurosciences, Sapienza University of Rome, Viale dell'Università 30, 00185, Rome (RM), Italy; IRCCS Neuromed, Via Atinense 18, 86077, Pozzilli (IS), Italy
| | - Anna Ladogana
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Anna Poleggi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
3
|
Emmi A, Mammana A, Sandre M, Baiardi S, Weis L, Rossi M, Magliocchetti F, Savarino E, Russo FP, Porzionato A, Carecchio M, Campagnolo M, Antonini A, Parchi P. Alpha-synuclein RT-QuIC assay in gastroduodenal and skin biopsies of Parkinson disease patients. Ann Clin Transl Neurol 2024. [PMID: 39708331 DOI: 10.1002/acn3.52282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/31/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
In this study, we compared the value of pathological alpha-synuclein (αSyn) seed amplification assay (SAA) in gastric and duodenal biopsies with skin biopsies in Parkinson disease (PD) patients with different disease duration. The accuracy of αSyn SAA was 87.7% in skin, 67.4% in duodenum, and 80.0% in gastric biopsies, with significantly higher sensitivity in advanced PD (skin: 81.8%; gastric: 88.9%; duodenal 58.8%). Misfolded αSyn was detected with higher sensitivity in advanced PD across all matrices, likely reflecting the progression of αSyn pathology. The seeding activity was lower in the duodenal than in the gastric wall, indicating differences in αSyn burden.
Collapse
Affiliation(s)
- Aron Emmi
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN-RND), University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Michele Sandre
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN-RND), University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Weis
- Institute of Hospitalization and Care of a Scientific Nature (IRCCS), IRCCS San Camillo Hospital, Venezia, Italy
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | | | - Edoardo Savarino
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Andrea Porzionato
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN-RND), University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Marta Campagnolo
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN-RND), University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Department of Neuroscience, Centre for Rare Neurological Diseases (ERN-RND), University of Padova, Padova, Italy
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Iftikhar IH, AlShimemeri S, Rabah H, Rao ST, BaHammam AS. Alpha-synuclein pathology in isolated rapid eye movement sleep behaviour disorder: a meta-analysis. J Sleep Res 2024; 33:e14204. [PMID: 38586895 DOI: 10.1111/jsr.14204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 04/09/2024]
Abstract
Accumulating evidence indicates that patients with isolated rapid eye movement sleep behaviour disorder (iRBD), a prodromal stage of synucleinopathies, show abnormal deposition of misfolded alpha-synuclein (a-Syn) in peripheral tissues. The clinical utility of testing for a-Syn in iRBD is unclear. This meta-analysis focused on the utility of testing for the abnormal a-Syn phosphorylated at Ser129 (p-syn) and a-Syn seeding activity (a-Syn seed amplification assays [aSyn-SAA]). Following an electronic database search, 15 studies were included that provided at a minimum data on test positivity in participants with iRBD. Test positivity from cerebrospinal fluid (CSF) was 80% (95% confidence interval [CI] 68-88%, I2 = 71%) and for skin was 74.8% (95% CI 53.2-88.5%, I2 = 64%) for aSyn-SAA and 78.5% (95% CI 70.4-84.9%, I2 = 14%) for p-syn. The phenoconversion rate ratio of biopsy-positive versus biopsy-negative iRBD was 1.28 (95% CI 0.68-2.41, I2 = 0%). Skin as a source had a specificity of 99% (95% CI 95-100%, I2 = 0%; p = 0.01 compared to CSF). As a test, p-syn, had a specificity of 100% (95% CI 93-100%, I2 = 0%; p < 0.001) compared to aSyn-SAA. The odds ratio of a-Syn test positivity in iRBD versus other RBDs was 112 (95% CI 20-629, I2 = 0%). These results demonstrate clinically significant test positivity in iRBD and favour skin over CSF as the source of a-Syn pathological analysis, and p-syn over aSyn-SAA as the testing method. Overall, these findings indicate that testing for a-Syn could help in differentiating iRBD from RBD secondary to other conditions.
Collapse
Affiliation(s)
- Imran H Iftikhar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, USA
| | - Sohaila AlShimemeri
- Neurology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hussein Rabah
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Saad Tauheed Rao
- Shifa College of Medicine (medical student), Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Ahmed S BaHammam
- Department of Medicine, University Sleep Disorders Center, and Pulmonary Service, King Saud University, Riyadh, Saudi Arabia
- Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Mayà G, Iranzo A, Gaig C, Sánchez-Valle R, Serradell M, Molina-Porcel L, Santamaria J, Gelpi E, Aldecoa I. Post-mortem neuropathology of idiopathic rapid eye movement sleep behaviour disorder: a case series. Lancet Neurol 2024; 23:1238-1251. [PMID: 39577924 DOI: 10.1016/s1474-4422(24)00402-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/28/2024] [Accepted: 09/24/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Idiopathic rapid eye movement (REM) sleep behaviour disorder (IRBD) is thought to be an early stage of α-synuclein-related neurodegenerative diseases. Nevertheless, the definitive identification of its biological substrate can be determined only by post-mortem neuropathology. We aimed to describe the post-mortem neuropathology of individuals with IRBD who developed or did not develop a neurodegenerative disease before death. METHODS In this case series at the Hospital Clinic de Barcelona, Barcelona, Spain, we examined post-mortem brain tissue and spinal cords from individuals diagnosed with IRBD by video polysomnography who became donors to the Neurological Tissue Bank between May 28, 2005, and March 23, 2023. We performed post-mortem neuropathology to assess the presence and distribution of neuronal loss, gliosis, and protein aggregates using antibodies against α-synuclein, amyloid β, phosphorylated tau, three-repeat and four-repeat tau isoforms, and TDP-43. Comparative statistical analyses were not done because of the small sample size, but differences observed across the nuclei and brain structures were described. FINDINGS The brains and spinal cords of 20 individuals with IRBD were examined (19 [95%] men, one [5%] woman). Their clinical antemortem diagnoses were of IRBD without any other neurological disorder in three (15%), Parkinson's disease without dementia in two (10%), Parkinson's disease dementia (PDD) in three (15%), and dementia with Lewy bodies (DLB) in 12 (60%) individuals. Post-mortem neuropathological diagnoses were Lewy body disease in 19 (95%) and multiple system atrophy (MSA) in one (5%). All participants with Lewy body disease and MSA showed neuronal loss, gliosis, and α-synuclein deposits in neurons and astrocytes. In all participants, α-synuclein was found in the structures that regulate REM sleep atonia (eg, subcoeruleus nucleus, gigantocellular reticular nucleus, laterodorsal tegmentum, and amygdala). Coexistent pathologies were found in all participants, including Alzheimer's disease pathology (amyloid β plaques and neurofibrillary tangles) in 14 (70%), ageing-related tau astrogliopathy in 12 (60%), cerebral amyloid angiopathy in 11 (55%), argyrophilic grain disease in four (20%), limbic-predominant age-related TDP-43 encephalopathy in four (20%), and early changes indicative of progressive supranuclear palsy in three (15%). In individuals with IRBD without any other neurological disorder and in those who developed Parkinson's disease without dementia, α-synuclein was found in the brainstem and limbic system and rarely in the cortex, whereas coexisting proteinopathies were few and showed mild pathological burden. In contrast, in individuals who developed PDD or DLB, α-synuclein had diffuse distribution in the brainstem, limbic system, and cortex, and multiple comorbid pathologies were common, particularly those related to Alzheimer's disease. INTERPRETATION Although limited by a relatively small sample size, our observations provide strong neuropathological evidence that IRBD is an early stage of α-synuclein-related neurodegenerative disease. Concomitant pathologies are frequent and their role remains to be clarified: some might have contributed to the development of dementia, but some might be age-related changes. Our findings could inform the design of clinical trials of compounds that target specific pathological proteins (eg, α-synuclein and amyloid β) in people with IRBD. FUNDING Fundación BBVA-Hospital Clínic de Barcelona.
Collapse
Affiliation(s)
- Gerard Mayà
- Sleep Unit, Neurology Service, FRCB-IDIBAPS, CIBERNED CB06/05/0018-ISCIII, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Alex Iranzo
- Sleep Unit, Neurology Service, FRCB-IDIBAPS, CIBERNED CB06/05/0018-ISCIII, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.
| | - Carles Gaig
- Sleep Unit, Neurology Service, FRCB-IDIBAPS, CIBERNED CB06/05/0018-ISCIII, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, FRCB-IDIBAPS, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Monica Serradell
- Sleep Unit, Neurology Service, FRCB-IDIBAPS, CIBERNED CB06/05/0018-ISCIII, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Laura Molina-Porcel
- Neurological Tissue Bank of the Biobank, FRCB-IDIBAPS, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Joan Santamaria
- Sleep Unit, Neurology Service, FRCB-IDIBAPS, CIBERNED CB06/05/0018-ISCIII, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Iban Aldecoa
- Neurological Tissue Bank of the Biobank, FRCB-IDIBAPS, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain; Department of Pathology, Biomedical Diagnostic Center (CDB), Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Galvin JE. Lewy Body Dementia. Continuum (Minneap Minn) 2024; 30:1673-1698. [PMID: 39620839 DOI: 10.1212/con.0000000000001496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Lewy body dementia (LBD) is an umbrella term describing two closely related conditions: Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB). LBD is the second most common cause of neurodegenerative dementia but is often underrecognized in clinical practice. This review covers the key epidemiologic, clinical, cognitive, behavioral, and biomarker features of LBD and discusses current treatment options. LATEST DEVELOPMENTS Indicative biomarkers of LBD improve the ability to make a diagnosis and include single-photon emission computed tomography (SPECT) of the dopamine system (brain) and the noradrenergic system (cardiac), and polysomnography. α-Synuclein-specific biomarkers in spinal fluid, skin, plasma, and brain imaging are in active development with some available for clinical use. Prodromal stages of PDD and DLB have been contextualized, and diagnostic criteria have been published. An emerging theme is whether an integrated staging system focusing on protein aggregation, rather than clinical symptoms, may advance research efforts. ESSENTIAL POINTS LBD is a common cause of cognitive impairment in older adults but is often subject to significant delays in diagnosis and treatment, increasing the burden on patients and family care partners. Understanding key features of disease and the use of biomarkers will improve recognition. Earlier detection may also facilitate the development of new therapeutics and enrollment in clinical trials.
Collapse
|
7
|
Stocchi F, Bravi D, Emmi A, Antonini A. Parkinson disease therapy: current strategies and future research priorities. Nat Rev Neurol 2024; 20:695-707. [PMID: 39496848 DOI: 10.1038/s41582-024-01034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/06/2024]
Abstract
Parkinson disease (PD) is the fastest growing neurological disorder globally and poses substantial management challenges owing to progressive disability, emergence of levodopa-resistant symptoms, and treatment-related complications. In this Review, we examine the current state of research into PD therapies and outline future priorities for advancing our understanding and treatment of the disease. We identify two main research priorities for the coming years: first, slowing the progression of the disease through the integration of sensitive biomarkers and targeted biological therapies, and second, enhancing existing symptomatic treatments, encompassing surgical and infusion therapies, with the goal of postponing complications and improving long-term patient management. The path towards disease modification is impeded by the multifaceted pathophysiology and diverse mechanisms underlying PD. Ongoing studies are directed at α-synuclein aggregation, complemented by efforts to address specific pathways associated with the less common genetic forms of the disease. The success of these efforts relies on establishing robust end points, incorporating technology, and identifying reliable biomarkers for early diagnosis and continuous monitoring of disease progression. In the context of symptomatic treatment, the focus should shift towards refining existing approaches and fostering the development of novel therapeutic strategies that target levodopa-resistant symptoms and clinical manifestations that substantially impair quality of life.
Collapse
Affiliation(s)
- Fabrizio Stocchi
- Department of Neurology, University San Raffaele, Rome, Italy.
- Deptartment of Neurology, Institute for Research and Medical Care, IRCCS San Raffaele, Rome, Italy.
| | - Daniele Bravi
- Deptartment of Neurology, Institute for Research and Medical Care, IRCCS San Raffaele, Rome, Italy
| | - Aron Emmi
- Center for Neurodegenerative Diseases (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
| | - Angelo Antonini
- Center for Neurodegenerative Diseases (CESNE), Department of Neuroscience, University of Padova, Padova, Italy
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, Padua Neuroscience Center (PNC), University of Padova, Padova, Italy
| |
Collapse
|
8
|
Wang Z, Wu L, Gerasimenko M, Gilliland T, Shah ZSA, Lomax E, Yang Y, Gunzler SA, Donadio V, Liguori R, Xu B, Zou WQ. Seeding activity of skin misfolded tau as a biomarker for tauopathies. Mol Neurodegener 2024; 19:92. [PMID: 39609917 PMCID: PMC11606191 DOI: 10.1186/s13024-024-00781-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Tauopathies are a group of age-related neurodegenerative diseases characterized by the accumulation of pathologically hyperphosphorylated tau protein in the brain, leading to prion-like aggregation and propagation. They include Alzheimer's disease (AD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD). Currently, reliable diagnostic biomarkers that directly reflect the capability of propagation and spreading of misfolded tau aggregates in peripheral tissues and body fluids are lacking. METHODS We utilized the seed-amplification assay (SAA) employing ultrasensitive real-time quaking-induced conversion (RT-QuIC) to assess the prion-like seeding activity of pathological tau in the skin of cadavers with neuropathologically confirmed tauopathies, including AD, PSP, CBD, and PiD, compared to normal controls. RESULTS We found that the skin tau-SAA demonstrated a significantly higher sensitivity (75-80%) and specificity (95-100%) for detecting tauopathy, depending on the tau substrates used. Moreover, the increased tau-seeding activity was also observed in biopsy skin samples from living AD and PSP patients examined. Analysis of the end products of skin-tau SAA confirmed that the increased seeding activity was accompanied by the formation of tau aggregates with different physicochemical properties related to two different tau substrates used. CONCLUSIONS Overall, our study provides proof-of-concept that the skin tau-SAA can differentiate tauopathies from normal controls, suggesting that the seeding activity of misfolded tau in the skin could serve as a diagnostic biomarker for tauopathies.
Collapse
Affiliation(s)
- Zerui Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Ling Wu
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, USA
| | - Maria Gerasimenko
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Tricia Gilliland
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zahid Syed Ali Shah
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Evalynn Lomax
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yirong Yang
- Institute of Neurology, Department of Neurology, Jiangxi Academy of Clinical Medical Sciences, Rare Disease Center, Key Laboratory of Rare Neurological Diseases of Jiangxi Province Health Commission, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Steven A Gunzler
- Neurological Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences of Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Rocco Liguori
- IRCCS Institute of Neurological Sciences of Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Bin Xu
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC, USA.
| | - Wen-Quan Zou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Institute of Neurology, Department of Neurology, Jiangxi Academy of Clinical Medical Sciences, Rare Disease Center, Key Laboratory of Rare Neurological Diseases of Jiangxi Province Health Commission, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
9
|
Kuang Y, Mao H, Huang X, Chen M, Dai W, Gan T, Wang J, Sun H, Lin H, Liu Q, Yang X, Xu PY. α-Synuclein seeding amplification assays for diagnosing synucleinopathies: an innovative tool in clinical implementation. Transl Neurodegener 2024; 13:56. [PMID: 39574205 PMCID: PMC11580393 DOI: 10.1186/s40035-024-00449-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/25/2024] [Indexed: 11/25/2024] Open
Abstract
The spectrum of synucleinopathies, including Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), is characterized by α-synuclein (αSyn) pathology, which serves as the definitive diagnostic marker. However, current diagnostic methods primarily rely on motor symptoms that manifest years after the initial neuropathological changes, thereby delaying potential treatment. The symptomatic overlap between PD and MSA further complicates the diagnosis, highlighting the need for precise and differential diagnostic methods for these overlapping neurodegenerative diseases. αSyn misfolding and aggregation occur before clinical symptoms appear, suggesting that detection of pathological αSyn could enable early molecular diagnosis of synucleinopathies. Recent advances in seed amplification assay (SAA) offer a tool for detecting neurodegenerative diseases by identifying αSyn misfolding in fluid and tissue samples, even at preclinical stages. Extensive research has validated the effectiveness and reproducibility of SAAs for diagnosing synucleinopathies, with ongoing efforts focusing on optimizing conditions for detecting pathological αSyn in more accessible samples and identifying specific αSyn species to differentiate between various synucleinopathies. This review offers a thorough overview of SAA technology, exploring its applications for diagnosing synucleinopathies, addressing the current challenges, and outlining future directions for its clinical use.
Collapse
Affiliation(s)
- Yaoyun Kuang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hengxu Mao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaoyun Huang
- Houjie Hospital of Dongguan, Dongguan, 523000, China
| | - Minshan Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei Dai
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, 830054, Xinjiang, China
| | - Tingting Gan
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Jiaqi Wang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Sun
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hao Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qin Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xinling Yang
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
10
|
Barba L, Abu-Rumeileh S, Barthel H, Massa F, Foschi M, Bellomo G, Gaetani L, Thal DR, Parnetti L, Otto M. Clinical and diagnostic implications of Alzheimer's disease copathology in Lewy body disease. Brain 2024; 147:3325-3343. [PMID: 38991041 DOI: 10.1093/brain/awae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024] Open
Abstract
Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with a higher risk of dementia and, consequently, be not rarely misdiagnosed. In this review, we summarize the current understanding of LBD-AD by discussing the synergistic effects of AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment for use in LBD-AD and their possible diagnostic and prognostic values. AD pathology can be predicted in vivo by means of CSF, MRI and PET markers, whereas the most promising technique to date for identifying Lewy pathology in different biological tissues is the α-synuclein seed amplification assay. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity as in pure AD. Implementing the use of blood-based AD biomarkers might allow faster screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account when considering differential diagnoses of dementia syndromes, to allow prognostic evaluation on an individual level, and to guide symptomatic and disease-modifying therapies.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig 04103, Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Matteo Foschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna 48121, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Dietmar R Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Leuven 3001, Belgium
- Department of Pathology, UZ Leuven, Leuven 3000, Belgium
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| |
Collapse
|
11
|
Gaetani L, Paolini Paoletti F, Mechelli A, Bellomo G, Parnetti L. Research advancement in fluid biomarkers for Parkinson's disease. Expert Rev Mol Diagn 2024; 24:885-898. [PMID: 39262126 DOI: 10.1080/14737159.2024.2403073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/07/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Diagnostic criteria for Parkinson's disease (PD) rely on clinical, mainly motor, features, implying that pre-motor phase cannot be accurately identified. To achieve a reliable early diagnosis, similar to what has been done for Alzheimer's disease (AD), a shift from clinical to biological identification of PD is being pursued. This shift has taken great advantage from the research on cerebrospinal fluid (CSF) biomarkers as they mirror the ongoing molecular pathogenic mechanisms taking place in PD, thus intercepting the disease timely with respect to clinical manifestations. AREAS COVERED CSF α-synuclein seed amplification assay (αS-SAA) has emerged as the most promising biomarker of α-synucleinopathy. CSF biomarkers reflecting AD-pathology and axonal damage (neurofilament light chain) and a novel marker of dopaminergic dysfunction (DOPA decarboxylase) add valuable diagnostic and prognostic information in the neurochemical characterization of PD. EXPERT OPINION A biological classification system of PD, encompassing pathophysiological and staging biomarkers, might ensure both early identification and prognostic characterization of the patients. This approach could allow for the best setting for disease-modifying treatments which are currently under investigation.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Alessandro Mechelli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
12
|
Campagnolo M, Weis L, Sandre M, Tushevski A, Russo FP, Savarino E, Carecchio M, Stocco E, Macchi V, De Caro R, Parchi P, Bubacco L, Porzionato A, Antonini A, Emmi A. Immune landscape of the enteric nervous system differentiates Parkinson's disease patients from controls: The PADUA-CESNE cohort. Neurobiol Dis 2024; 200:106609. [PMID: 39048026 DOI: 10.1016/j.nbd.2024.106609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Gastrointestinal dysfunction has emerged as a prominent early feature of Parkinson's Disease, shedding new light on the pivotal role of the enteric nervous system in its pathophysiology. However, the role of immune-cell clusters and inflammatory and glial markers in the gut pathogenetic process needs further elucidation. OBJECTIVES We aimed to study duodenum tissue samples to characterize PD's enteric nervous system pathology further. Twenty patients with advanced PD, six with early PD, and 18 matched controls were included in the PADUA-CESNE cohort. METHODS Duodenal biopsies from 26 patients with early to advanced stage PD and 18 age-matched HCs were evaluated for the presence of surface markers (CD3+, CD4+, CD8+, CD20+, CD68+, HLA-DR), presence of misfolded alpha-synuclein and enteric glial alteration (GFAP). Correlation of immulogic pattern and clinical characteristic were analyzed. RESULTS The findings validate that in patients with Parkinson's Disease, the activation and reactive gliosis are linked to the neurodegeneration triggered by the presence of misfolded alpha-synuclein in the enteric nervous system. This process intensifies from the initial to the advanced stages of the disease. The clusters of T- and B-lymphocytes in the enteric system, along with the overall expression of HLA-DR in antigen-presenting cells, exceeded those in the control group. Conversely, no differences in terms of macrophage populations were found. CONCLUSIONS These findings broaden our understanding of the mechanisms underlying the enteric nervous system's involvement in PD and point to the gastrointestinal system as a potential therapeutic target, especially in the early stages of the disease. Moreover, our results propose a role of T- and B-lymphocytes in maintaining inflammation and ultimately influencing alpha-synuclein misfolding and aggregation.
Collapse
Affiliation(s)
- Marta Campagnolo
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Luca Weis
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy
| | - Michele Sandre
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy
| | - Aleksandar Tushevski
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy
| | - Francesco Paolo Russo
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Edoardo Savarino
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy
| | - Miryam Carecchio
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy
| | - Elena Stocco
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Padova, Italy; Department of Women's and Children's Health, University of Padova, Padova, Italy; Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
| | - Veronica Macchi
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy.
| | - Raffaele De Caro
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luigi Bubacco
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Department of Biology, University of Padova, Padova, Italy
| | - Andrea Porzionato
- Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy
| | - Angelo Antonini
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Padua Neuroscience Center (PNC), University of Padova, Padova, Italy
| | - Aron Emmi
- Parkinson and Movement Disorders Unit, Centre for Rare Neurological Diseases (ERN-RND), Department of Neuroscience, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), University of Padova, Padova, Italy; Institute of Human Anatomy, Department of Neuroscience, University of Padova, Padova, Italy; Padua Neuroscience Center (PNC), University of Padova, Padova, Italy.
| |
Collapse
|
13
|
Mammana A, Baiardi S, Rossi M, Quadalti C, Ticca A, Magliocchetti F, Bernhardt A, Capellari S, Parchi P. Improving protocols for α-synuclein seed amplification assays: analysis of preanalytical and analytical variables and identification of candidate parameters for seed quantification. Clin Chem Lab Med 2024; 62:2001-2010. [PMID: 38456740 DOI: 10.1515/cclm-2023-1472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVES The effect of preanalytical and analytical factors on the α-synuclein (α-syn) seed amplification assay's (SAA) performance has not been fully explored. Similarly, there is limited knowledge about the most suitable assay protocol and kinetic parameters for misfolded α-syn seed quantification. METHODS We studied the effect of centrifugation, repeated freeze-thaw cycles (up to seven), delayed freezing, detergent addition, and blood contamination on the performance of the cerebrospinal fluid (CSF) α-syn SAA real-time quaking-induced conversion (RT-QuIC). Moreover, we analysed the inter- and intra-plate variability, the recombinant protein batch effect, and the RT-QuIC parameters' variability when multiple samples were run in controlled conditions. Finally, we evaluated the assay potential of quantifying α-syn seed by assessing kinetic curves in serial CSF dilutions. RESULTS Among tested preanalytical variables, a ≥0.01 % blood contamination and adding detergents significantly affected the RT-QuIC kinetic parameters and the number of positive replicates. Increasing the number of replicates improved result reproducibility. The number of positive replicates in serially diluted CSF samples improved discrimination between samples with high and low seeding activity, and the time to threshold (LAG) was the most reliable kinetic parameter in multiple experiment settings. CONCLUSIONS Preanalytical variables affecting α-syn RT-QuIC performance are limited to blood contamination and detergent addition. The number of positive replicates and the LAG are the most reliable variables for quantifying α-syn seeding activity. Their consistent measurement in serial dilution experiments, especially when associated with an increased number of sample replicates, will help to develop the α-syn RT-QuIC assay further into a quantitative test.
Collapse
Affiliation(s)
- Angela Mammana
- 419170 IRCCS Istituto delle Scienze Neurologiche di Bologna , Bologna, Italy
| | - Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, 9296 University of Bologna , Bologna, Italy
| | - Marcello Rossi
- 419170 IRCCS Istituto delle Scienze Neurologiche di Bologna , Bologna, Italy
| | - Corinne Quadalti
- 419170 IRCCS Istituto delle Scienze Neurologiche di Bologna , Bologna, Italy
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, 9296 University of Bologna , Bologna, Italy
| | | | - Alexander Bernhardt
- Department of Neurology, 9183 Ludwig-Maximilians-Universität München , Munich, Germany
| | - Sabina Capellari
- 419170 IRCCS Istituto delle Scienze Neurologiche di Bologna , Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, 9296 University of Bologna , Bologna, Italy
| | - Piero Parchi
- 419170 IRCCS Istituto delle Scienze Neurologiche di Bologna , Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, 9296 University of Bologna , Bologna, Italy
| |
Collapse
|
14
|
Zhou Y, Liu X, Xu B. Research Progress on the Relationship between Parkinson's Disease and REM Sleep Behavior Disorder. J Integr Neurosci 2024; 23:166. [PMID: 39344226 DOI: 10.31083/j.jin2309166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 10/01/2024] Open
Abstract
An individual's quality of life is greatly affected by Parkinson's disease (PD), a prevalent neurological degenerative condition. Rapid eye movement (REM) sleep behavior disorder (RBD) is a prominent non-motor symptom commonly associated with PD. Previous studies have shown a close relationship between PD and RBD. In addition to being a prodromal symptom of PD, RBD has a major negative impact on the prognosis of PD patients. This intrinsic connection indicates that there is a bidirectional relationship between PD and RBD. This paper provides a comprehensive review of the pathological mechanism related to PD and RBD, including the α-synuclein pathological deposition, abnormal iron metabolism, neuroinflammation, glymphatic system dysfunction and dysbiosis of the gut microbiota. Increasing evidence has shown that RBD patients have the same pathogenic mechanisms that underlie PD, but relatively little research has been done on how RBD contributes to PD progression. Therefore, a more thorough investigation is warranted to characterise how RBD affects the course of PD, in order to prepare for future therapeutic trials.
Collapse
Affiliation(s)
- Yu Zhou
- The Second Clinical Medical College of Zhejiang Chinese Medical University, 310000 Hangzhou, Zhejiang, China
| | - Xiaoli Liu
- Department of Neurology, Zhejiang Hospital Affiliated to Zhejiang University, 310000 Hangzhou, Zhejiang, China
| | - Bin Xu
- Department of Neurology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, 310000 Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Sade O, Fischel D, Barak-Broner N, Halevi S, Gottfried I, Bar-On D, Sachs S, Mirelman A, Thaler A, Gour A, Kestenbaum M, Gana Weisz M, Anis S, Soto C, Roitman MS, Shahar S, Doppler K, Sauer M, Giladi N, Lev N, Alcalay RN, Hassin-Baer S, Ashery U. A novel super-resolution microscopy platform for cutaneous alpha-synuclein detection in Parkinson's disease. Front Mol Neurosci 2024; 17:1431549. [PMID: 39296283 PMCID: PMC11409901 DOI: 10.3389/fnmol.2024.1431549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/26/2024] [Indexed: 09/21/2024] Open
Abstract
Alpha-synuclein (aSyn) aggregates in the central nervous system are the main pathological hallmark of Parkinson's disease (PD). ASyn aggregates have also been detected in many peripheral tissues, including the skin, thus providing a novel and accessible target tissue for the detection of PD pathology. Still, a well-established validated quantitative biomarker for early diagnosis of PD that also allows for tracking of disease progression remains lacking. The main goal of this research was to characterize aSyn aggregates in skin biopsies as a comparative and quantitative measure for PD pathology. Using direct stochastic optical reconstruction microscopy (dSTORM) and computational tools, we imaged total and phosphorylated-aSyn at the single molecule level in sweat glands and nerve bundles of skin biopsies from healthy controls (HCs) and PD patients. We developed a user-friendly analysis platform that offers a comprehensive toolkit for researchers that combines analysis algorithms and applies a series of cluster analysis algorithms (i.e., DBSCAN and FOCAL) onto dSTORM images. Using this platform, we found a significant decrease in the ratio of the numbers of neuronal marker molecules to phosphorylated-aSyn molecules, suggesting the existence of damaged nerve cells in fibers highly enriched with phosphorylated-aSyn molecules. Furthermore, our analysis found a higher number of aSyn aggregates in PD subjects than in HC subjects, with differences in aggregate size, density, and number of molecules per aggregate. On average, aSyn aggregate radii ranged between 40 and 200 nm and presented an average density of 0.001-0.1 molecules/nm2. Our dSTORM analysis thus highlights the potential of our platform for identifying quantitative characteristics of aSyn distribution in skin biopsies not previously described for PD patients while offering valuable insight into PD pathology by elucidating patient aSyn aggregation status.
Collapse
Affiliation(s)
- Ofir Sade
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Daphna Fischel
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noa Barak-Broner
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Shir Halevi
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Gottfried
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Dana Bar-On
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Stefan Sachs
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Anat Mirelman
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Avner Thaler
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Aviv Gour
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Meir Medical Center, Kfar Saba, Israel
| | - Meir Kestenbaum
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Meir Medical Center, Kfar Saba, Israel
| | - Mali Gana Weisz
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Saar Anis
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Movement Disorders Institute, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, University of Texas Medical School, Houston, TX, United States
| | - Melanie Shanie Roitman
- Department of Neurology, Movement Disorders Institute, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Shimon Shahar
- Department of Statistics, Exact Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nir Giladi
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nirit Lev
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Meir Medical Center, Kfar Saba, Israel
| | - Roy N Alcalay
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Hassin-Baer
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurology, Movement Disorders Institute, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry, Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Iranzo A, Cochen De Cock V, Fantini ML, Pérez-Carbonell L, Trotti LM. Sleep and sleep disorders in people with Parkinson's disease. Lancet Neurol 2024; 23:925-937. [PMID: 38942041 DOI: 10.1016/s1474-4422(24)00170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/02/2024] [Accepted: 04/16/2024] [Indexed: 06/30/2024]
Abstract
Sleep disorders are common in people with Parkinson's disease. These disorders, which increase in frequency throughout the course of the neurodegenerative disease and impair quality of life, include insomnia, excessive daytime sleepiness, circadian disorders, obstructive sleep apnoea, restless legs syndrome, and rapid eye movement (REM) sleep behaviour disorder. The causes of these sleep disorders are complex and multifactorial, including the degeneration of the neural structures that modulate sleep, the detrimental effect of some medications on sleep, the parkinsonian symptoms that interfere with mobility and comfort in bed, and comorbidities that disrupt sleep quality and quantity. The clinical evaluation of sleep disorders include both subjective (eg, questionnaires or diaries) and objective (eg, actigraphy or video polysomnography) assessments. The management of patients with Parkinson's disease and a sleep disorder is challenging and should be individualised. Treatment can include education aiming at changes in behaviour (ie, sleep hygiene), cognitive behavioural therapy, continuous dopaminergic stimulation at night, and specific medications. REM sleep behaviour disorder can occur several years before the onset of parkinsonism, suggesting that the implementation of trials of neuroprotective therapies should focus on people with this sleep disorder.
Collapse
Affiliation(s)
- Alex Iranzo
- Sleep Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain; IDIBAPS, Universitat de Barcelona, Barcelona, Spain; CIBERNED, Universitat de Barcelona, Barcelona, Spain.
| | - Valerie Cochen De Cock
- Sleep and Neurology Department, Beau Soleil Clinic, Montpellier, France; EuroMov Digital Health in Motion, University of Montpellier, IMT Mines Ales, Montpellier, France
| | - María Livia Fantini
- Neurophysiology Unit, Neurology Department, Université Clermont Auvergne, CNRS, Institut Pascal, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Laura Pérez-Carbonell
- Sleep Disorders Centre, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | - Lynn Marie Trotti
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Emory Sleep Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
17
|
Muñoz-Lopetegi A, Baiardi S, Balasa M, Mammana A, Mayà G, Rossi M, Serradell M, Zenesini C, Ticca A, Santamaria J, Dellavalle S, Gaig C, Iranzo A, Parchi P. CSF markers of neurodegeneration Alzheimer's and Lewy body pathology in isolated REM sleep behavior disorder. NPJ Parkinsons Dis 2024; 10:157. [PMID: 39147825 PMCID: PMC11327307 DOI: 10.1038/s41531-024-00770-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024] Open
Abstract
We investigated the biomarker profile of neurodegeneration, Alzheimer's and Lewy body pathology in the CSF of 148 polysomnography-confirmed patients with isolated REM sleep behavior disorder (IRBD), a condition that precedes Parkinson's disease (PD) and dementia with Lewy bodies (DLB). We assessed misfolded α-synuclein (AS) by RT-QuIC assay, amyloid-beta peptides (Aβ42 and Aβ40), phosphorylated tau (p-tau), and total tau (t-tau) by CLEIA and neurofilament light chain (NfL) by ELISA. We detected AS in 75.3% of patients, pathologically decreased Aβ42/Aβ40 ratio in 22.5%, increased p-tau in 15.5%, increased t-tau in 14.9%, and elevated NfL in 14.7%. After a mean follow-up of 2.48 ± 2.75 years, 47 (38.1%) patients developed PD (n = 24) or DLB (n = 23). At CSF collection, AS positivity [HR 4.05 (1.26-12.99), p = 0.019], mild cognitive impairment [3.86 (1.96-7.61), p < 0.001], and abnormal DAT-SPECT [2.31 (1.09-4.91), p < 0.030] were independent predictors of conversion to PD and DLB. Among the other CSF markers, only elevated p-tau/Aβ42 was predictive of conversion, although only to DLB and not as an independent variable. In IRBD, CSF AS assessment by RT-QuIC provides an added value in defining the risk of short-term conversion to PD and DLB independent of clinical and instrumental investigations. Positive Alzheimer's disease (AD) pathology markers and elevated NfL occur in a subgroup of patients, but p-tau/Aβ42 is the only marker that predicts short-term conversion to DLB. Longer follow-up is needed to assess if AD biomarkers predict the later development of PD and DLB in IRBD.
Collapse
Affiliation(s)
- Amaia Muñoz-Lopetegi
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Simone Baiardi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic Barcelona, Barcelona, Spain
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Gerard Mayà
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Mónica Serradell
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Corrado Zenesini
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Joan Santamaria
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Sofia Dellavalle
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Carles Gaig
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Alex Iranzo
- Neurology Service, Sleep Unit, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain.
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
18
|
Coughlin DG, MacLeod KR, Middleton JS, Bozoki AC, Galvin JE, Irwin DJ, Lippa CF, Litvan I, Lopez OL, Berman S, Tsuang DW, Zabetian CP, Honig LS, Marder KS, Fleisher JE, Sabbagh M, Wint D, Taylor AS, Bekris L, Leverenz JB, Galasko D. Association of CSF α-Synuclein Seeding Amplification Assay Results With Clinical Features of Possible and Probable Dementia With Lewy Bodies. Neurology 2024; 103:e209656. [PMID: 39013126 PMCID: PMC11238940 DOI: 10.1212/wnl.0000000000209656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/28/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The clinical diagnosis of dementia with Lewy bodies (DLB) depends on identifying significant cognitive decline accompanied by core features of parkinsonism, visual hallucinations, cognitive fluctuations, and REM sleep behavior disorder (RBD). Hyposmia is one of the several supportive features. α-Synuclein seeding amplification assays (αSyn-SAAs) may enhance diagnostic accuracy by detecting pathologic αSyn seeds in CSF. In this study, we examine how different clinical features associate with CSF αSyn-SAA positivity in a large group of clinically diagnosed participants with DLB. METHODS Cross-sectional and longitudinal CSF samples from the multicentered observational cohort study of the DLB Consortium and similar studies within the Parkinson's Disease Biomarker Program, contributed by academic medical centers in the United States, underwent αSyn-SAA testing. Participants included those clinically diagnosed with DLB and 2 control cohorts. Associations between core DLB features and olfaction with αSyn-SAA positivity were evaluated using logistic regression. RESULTS CSF samples from 191 participants diagnosed with DLB (mean age 69.9 ± 6.8, 15% female), 50 age-matched and sex-matched clinical control participants, and 49 younger analytical control participants were analyzed. Seventy-two percent (137/191) of participants with DLB had positive αSyn-SAAs vs 4% of the control groups. Among participants with DLB, those who were αSyn-SAA-positive had lower Montreal Cognitive Assessment scores (18.8 ± 5.7 vs 21.2 ± 5.2, p = 0.01), had worse parkinsonism on the Movement Disorders Society Unified Parkinson's Disease Rating Scale part III (33.8 ± 15.1 vs 25.6 ± 16.4, p = 0.001), were more likely to report RBD (114/133 [86%] vs 33/53 [62%], p < 0.0001), and had worse hyposmia on the University of Pennsylvania Smell Identification Test (UPSIT) (94/105 [90%] below 15th percentile vs 14/44 [32%], p < 0.0001). UPSIT percentile had the highest area under the curve (0.87, 95% CI 0.81-0.94) in predicting αSyn-SAA positivity and participants scoring at or below the 15th percentile of age and sex normative values had 18.3 times higher odds (95% CI 7.52-44.6) of having a positive αSyn-SAA test. Among 82 participants with longitudinal CSF samples, 81 (99%) had the same αSyn-SAA result for initial and follow-up specimens. DISCUSSION A substantial proportion of clinically diagnosed participants with DLB had negative αSyn-SAA results. Hyposmia was the strongest clinical predictor of αSyn-SAA positivity. Hyposmia and αSyn-SAA may have utility in improving the diagnostic assessment of individuals with potential DLB. CLASSIFICATION OF EVIDENCE This study provided Class III evidence that CSF αSyn-SAA distinguishes patients with clinically diagnosed DLB from normal controls.
Collapse
Affiliation(s)
- David G Coughlin
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Karen R MacLeod
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - John S Middleton
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Andrea C Bozoki
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - James E Galvin
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - David J Irwin
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Carol F Lippa
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Irene Litvan
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Oscar L Lopez
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Sarah Berman
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Debby W Tsuang
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Cyrus P Zabetian
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Lawrence S Honig
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Karen S Marder
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Jori E Fleisher
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Marwan Sabbagh
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Dylan Wint
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Angela S Taylor
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Lynn Bekris
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - James B Leverenz
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| | - Douglas Galasko
- From the Department of Neurosciences (D.G.C., I.L., D.G.), University of California San Diego; Clinical Laboratory (K.R.M., J.S.M.), Amprion Inc., La Jolla, CA; Department of Neurology (A.C.B.), University of North Carolina, Chapel Hill, NC; Department of Neurology (J.E.G.), University of Miami, FL; Department of Neurology (D.J.I.), University of Pennsylvania, Philadelphia; Department of Neurology (C.F.L.), Thomas Jefferson University, Philadelphia, PA; Department of Neurology (O.L.L., S.B.), University of Pittsburgh, PA; Department of Neurology (D.W.T., C.P.Z.), University of Washington and Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA; Department of Neurology (L.S.H., K.S.M.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (J.E.F.), Rush University, Chicago, IL; Department of Neurology (M.S.), Barrow Neurological Institute, AZ; Department of Neurology (D.W., L.B., J.B.L.), Cleveland Clinic, OH; and Lewy Body Dementia Association (A.S.T.), Lilburn, GA
| |
Collapse
|
19
|
Arnaldi D, Iranzo A, Nobili F, Postuma RB, Videnovic A. Developing disease-modifying interventions in idiopathic REM sleep behavior disorder and early synucleinopathy. Parkinsonism Relat Disord 2024; 125:107042. [PMID: 38943771 DOI: 10.1016/j.parkreldis.2024.107042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/01/2024]
Abstract
Alpha-synucleinopathies are prevalent neurological disorders that cause significant disability, leading to progressive clinical deterioration that is currently managed solely through symptomatic treatment. Efforts to evaluate disease-modifying therapies during the established stage of the disease have not yielded positive outcomes in terms of clinical or imaging efficacy endpoints. However, alpha-synucleinopathies have a long prodromal phase that presents a promising opportunity for intervention with disease-modifying therapies. The presence of polysomnography-confirmed REM sleep behavior disorder (RBD) is the most reliable risk factor for identifying individuals in the prodromal stage of alpha-synucleinopathy. This paper discusses the rationale behind targeting idiopathic/isolated RBD in disease-modifying trials and outlines possible study designs, including strategies for patient stratification, selection of biomarkers to assess disease progression and patient eligibility, as well as the identification of suitable endpoints. Additionally, the potential targets for disease-modifying treatment in alpha-synucleinopathies are summarized.
Collapse
Affiliation(s)
- Dario Arnaldi
- Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Alex Iranzo
- Neurology Service, Sleep Disorder Centre, Hospital Clinic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain
| | - Flavio Nobili
- Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Ronald B Postuma
- Department of Neurology, McGill University, Montreal Neurological Institute, Montreal, Canada; Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, Canada
| | - Aleksandar Videnovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Kluge A, Schaeffer E, Bunk J, Sommerauer M, Röttgen S, Schulte C, Roeben B, von Thaler AK, Welzel J, Lucius R, Heinzel S, Xiang W, Eschweiler GW, Maetzler W, Suenkel U, Berg D. Detecting Misfolded α-Synuclein in Blood Years before the Diagnosis of Parkinson's Disease. Mov Disord 2024; 39:1289-1299. [PMID: 38651526 DOI: 10.1002/mds.29766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Identifying individuals with Parkinson's disease (PD) already in the prodromal phase of the disease has become a priority objective for opening a window for early disease-modifying therapies. OBJECTIVE The aim was to evaluate a blood-based α-synuclein seed amplification assay (α-syn SAA) as a novel biomarker for diagnosing PD in the prodromal phase. METHODS In the TREND study (University of Tuebingen) biennial blood samples of n = 1201 individuals with/without increased risk for PD were taken prospectively over 4 to 10 years. We retrospectively analyzed blood samples of 12 participants later diagnosed with PD during the study to detect and amplify pathological α-syn conformers derived from neuronal extracellular vesicles using (1) immunoblot analyses with an antibody against these conformers and (2) an α-syn-SAA. Additionally, blood samples of n = 13 healthy individuals from the TREND cohort and n = 20 individuals with isolated rapid eye movement sleep behavior disorder (iRBD) from the University Hospital Cologne were analyzed. RESULTS All individuals with PD showed positive immunoblots and a positive α-syn SAA at the time of diagnosis. Moreover, all PD patients showed a positive α-syn SAA 1 to 10 years before clinical diagnosis. In the iRBD cohort, 30% showed a positive α-syn SAA. All healthy controls had a negative SAA. CONCLUSIONS We here demonstrate the possibility to detect and amplify pathological α-syn conformers in peripheral blood up to 10 years before the clinical diagnosis of PD in individuals with and without iRBD. The findings of this study indicate that this blood-based α-syn SAA assay has the potential to serve as a diagnostic biomarker for prodromal PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Josina Bunk
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Michael Sommerauer
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany
| | - Sinah Röttgen
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Benjamin Roeben
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Anna-Katharina von Thaler
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Julius Welzel
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Ralph Lucius
- Institute of Anatomy, Kiel University, Kiel, Germany
| | - Sebastian Heinzel
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Gerhard W Eschweiler
- Geriatric Center, University Hospital Tübingen, Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Ulrike Suenkel
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Partner Site Tübingen, Tübingen, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Kiel, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
21
|
Mao H, Kuang Y, Feng D, Chen X, Lu L, Xia W, Gan T, Huang W, Guo W, Yi H, Yang Y, Wu Z, Dai W, Sun H, Wu J, Zhang R, Zhang S, Lin X, Yong Y, Yang X, Li H, Wu W, Huang X, Bian Z, Wong HLX, Wang XL, Poppell M, Ren Y, Liu C, Zou WQ, Chen S, Xu PY. Ultrasensitive detection of aggregated α-synuclein using quiescent seed amplification assay for the diagnosis of Parkinson's disease. Transl Neurodegener 2024; 13:35. [PMID: 39049095 PMCID: PMC11267792 DOI: 10.1186/s40035-024-00426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Seed amplification assays (SAA) enable the amplification of pathological misfolded proteins, including α-synuclein (αSyn), in both tissue homogenates and body fluids of Parkinson's disease (PD) patients. SAA involves repeated cycles of shaking or sonication coupled with incubation periods. However, this amplification scheme has limitations in tracking protein propagation due to repeated fragmentation. METHODS We introduced a modified form of SAA, known as Quiescent SAA (QSAA), and evaluated biopsy and autopsy samples from individuals clinically diagnosed with PD and those without synucleinopathies (control group). Brain biopsy samples were obtained from 14 PD patients and 6 controls without synucleinopathies. Additionally, skin samples were collected from 214 PD patients and 208 control subjects. Data were analyzed from April 2019 to May 2023. RESULTS QSAA successfully amplified αSyn aggregates in brain tissue sections from mice inoculated with pre-formed fibrils. In the skin samples from 214 PD cases and 208 non-PD cases, QSAA demonstrated high sensitivity (90.2%) and specificity (91.4%) in differentiating between PD and non-PD cases. Notably, more αSyn aggregates were detected by QSAA compared to immunofluorescence with the pS129-αSyn antibody in consecutive slices of both brain and skin samples. CONCLUSION We introduced the new QSAA method tailored for in situ amplification of αSyn aggregates in brain and skin samples while maintaining tissue integrity, providing a streamlined approach to diagnosing PD with individual variability. The integration of seeding activities with the location of deposition of αSyn seeds advances our understanding of the mechanism underlying αSyn misfolding in PD.
Collapse
Affiliation(s)
- Hengxu Mao
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yaoyun Kuang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Du Feng
- School of Basic Medical Science, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiang Chen
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Lin Lu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Tingting Gan
- Deptartment of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Weimeng Huang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wenyuan Guo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hancun Yi
- Institute of Neurology, Jiangxi Academy of Medical Clinical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yirong Yang
- Institute of Neurology, Jiangxi Academy of Medical Clinical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhuohua Wu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei Dai
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Sun
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jieyuan Wu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Rui Zhang
- Deptartment of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiuli Lin
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yuxuan Yong
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xinling Yang
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Hongyan Li
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, 830054, China
| | - Wenjun Wu
- Department of Neurology, Zhongshan City People's Hospital, Zhongshan, 528400, China
| | - Xiaoyun Huang
- Dongguan Songshan Lake Central Hospital, Dongguan, 523000, China
| | - Zhaoxiang Bian
- Jockey Club School of Chinese Medicine, Baptist University Road, Hong Kong, 999077, China
| | - Hoi Leong Xavier Wong
- Jockey Club School of Chinese Medicine, Baptist University Road, Hong Kong, 999077, China
| | - Xin-Lu Wang
- Department of Nuclear Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Michael Poppell
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, 32306, USA
| | - Yi Ren
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, 32306, USA
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Wen-Quan Zou
- Institute of Neurology, Jiangxi Academy of Medical Clinical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Shengdi Chen
- Department of Neurology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ping-Yi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
22
|
Kuang Y, Mao H, Gan T, Guo W, Dai W, Huang W, Wu Z, Li H, Huang X, Yang X, Xu PY. A skin-specific α-Synuclein seeding amplification assay for diagnosing Parkinson's disease. NPJ Parkinsons Dis 2024; 10:129. [PMID: 38961119 PMCID: PMC11222486 DOI: 10.1038/s41531-024-00738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/31/2024] [Indexed: 07/05/2024] Open
Abstract
The seeding amplification assay (SAA) has recently emerged as a valuable tool for detecting α-synuclein (αSyn) aggregates in various clinically accessible biospecimens. Despite its efficiency and specificity, optimal tissue-specific conditions for distinguishing Parkinson's disease (PD) from non-PD outside the brain remain underexplored. This study systematically evaluated 150 reaction conditions to identify the one with the highest discriminatory potential between PD and non-synucleinopathy controls using skin samples, resulting in a modified SAA. The streamlined SAA achieved an overall sensitivity of 92.46% and specificity of 93.33% on biopsy skin samples from 332 PD patients and 285 controls within 24 h. Inter-laboratory reproducibility demonstrated a Cohen's kappa value of 0.87 (95% CI 0.69-1.00), indicating nearly perfect agreement. Additionally, αSyn seeds in the skin were stable at -80 °C but were vulnerable to short-term exposure to non-ultra-low temperatures and grinding. This study thoroughly investigated procedures for sample preprocessing, seed amplification, and storage, introducing a well-structured experimental framework for PD diagnosis using skin samples.
Collapse
Affiliation(s)
- Yaoyun Kuang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hengxu Mao
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Tingting Gan
- Department of Neurology, the First Affiliated Hospital of Gannan Medical University, 341000, Ganzhou, China
| | - Wenyuan Guo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei Dai
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, 830054, Urumqi, Xinjiang, China
| | - Weimeng Huang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhuohua Wu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hongyan Li
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, 830054, Urumqi, Xinjiang, China
| | - Xiaoyun Huang
- Dongguan Songshan Lake Central Hospital, 523000, Donggguan, China.
| | - Xinling Yang
- The Second Affiliated Hospital of Xinjiang Medical University, 830054, Urumqi, Xinjiang, China.
| | - Ping-Yi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
23
|
Soto C. α-Synuclein seed amplification technology for Parkinson's disease and related synucleinopathies. Trends Biotechnol 2024; 42:829-841. [PMID: 38395703 PMCID: PMC11223967 DOI: 10.1016/j.tibtech.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
Synucleinopathies are a group of neurodegenerative diseases (NDs) associated with cerebral accumulation of α-synuclein (αSyn) misfolded aggregates. At this time, there is no effective treatment to stop or slow down disease progression, which in part is due to the lack of an early and objective biochemical diagnosis. In the past 5 years, the seed amplification technology has emerged for highly sensitive identification of these diseases, even at the preclinical stage of the illness. Much research has been done in multiple laboratories to validate the efficacy and reproducibility of this assay. This article provides a comprehensive review of this technology, including its conceptual basis and its multiple applications for disease diagnosis, as well for understanding of the disease biology and therapeutic development.
Collapse
Affiliation(s)
- Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, TX77030, USA.
| |
Collapse
|
24
|
Boeve BF, Davis AA, Ju YE, Kantarci K, Singer W, Videnovic A. Concerns with the new biological research criteria for synucleinopathy. Lancet Neurol 2024; 23:659-660. [PMID: 38876735 DOI: 10.1016/s1474-4422(24)00213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/09/2024] [Indexed: 06/16/2024]
Affiliation(s)
- Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Albert A Davis
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Yo-El Ju
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Kejal Kantarci
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wolfgang Singer
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Aleks Videnovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Nardelli D, Gambioli F, De Bartolo MI, Mancinelli R, Biagioni F, Carotti S, Falato E, Leodori G, Puglisi-Allegra S, Vivacqua G, Fornai F. Pain in Parkinson's disease: a neuroanatomy-based approach. Brain Commun 2024; 6:fcae210. [PMID: 39130512 PMCID: PMC11311710 DOI: 10.1093/braincomms/fcae210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/23/2024] [Accepted: 06/17/2024] [Indexed: 08/13/2024] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder characterized by the deposition of misfolded alpha-synuclein in different regions of the central and peripheral nervous system. Motor impairment represents the signature clinical expression of Parkinson's disease. Nevertheless, non-motor symptoms are invariably present at different stages of the disease and constitute an important therapeutic challenge with a high impact for the patients' quality of life. Among non-motor symptoms, pain is frequently experienced by patients, being present in a range of 24-85% of Parkinson's disease population. Moreover, in more than 5% of patients, pain represents the first clinical manifestation, preceding by decades the exordium of motor symptoms. Pain implies a complex biopsychosocial experience with a downstream complex anatomical network involved in pain perception, modulation, and processing. Interestingly, all the anatomical areas involved in pain network can be affected by a-synuclein pathology, suggesting that pathophysiology of pain in Parkinson's disease encompasses a 'pain spectrum', involving different anatomical and neurochemical substrates. Here the various anatomical sites recruited in pain perception, modulation and processing are discussed, highlighting the consequences of their possible degeneration in course of Parkinson's disease. Starting from peripheral small fibres neuropathy and pathological alterations at the level of the posterior laminae of the spinal cord, we then describe the multifaceted role of noradrenaline and dopamine loss in driving dysregulated pain perception. Finally, we focus on the possible role of the intertwined circuits between amygdala, nucleus accumbens and habenula in determining the psycho-emotional, autonomic and cognitive experience of pain in Parkinson's disease. This narrative review provides the first anatomically driven comprehension of pain in Parkinson's disease, aiming at fostering new insights for personalized clinical diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Domiziana Nardelli
- Laboratory of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Roma, Rome 00128, Italy
| | - Francesco Gambioli
- Laboratory of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Roma, Rome 00128, Italy
| | | | - Romina Mancinelli
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Roma, Rome 00161, Italy
| | | | - Simone Carotti
- Laboratory of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Roma, Rome 00128, Italy
| | - Emma Falato
- Laboratory of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Roma, Rome 00128, Italy
| | - Giorgio Leodori
- IRCCS Neuromed, Pozzilli, IS 86077, Italy
- Department of Human Neuroscience, Sapienza University of Roma, Rome 00185, Italy
| | | | - Giorgio Vivacqua
- Laboratory of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Roma, Rome 00128, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Pozzilli, IS 86077, Italy
- Department of Experimental Morphology and Applied Biology, University of Pisa, Pisa 56122, Italy
| |
Collapse
|
26
|
Pérez-Carbonell L, Iranzo A. REM sleep and neurodegeneration. J Sleep Res 2024:e14263. [PMID: 38867555 DOI: 10.1111/jsr.14263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Several brainstem, subcortical and cortical areas are involved in the generation of rapid eye movement (REM) sleep. The alteration of these structures as a result of a neurodegenerative process may therefore lead to REM sleep anomalies. REM sleep behaviour disorder is associated with nightmares, dream-enacting behaviours and increased electromyographic activity in REM sleep. Its isolated form is a harbinger of synucleinopathies such as Parkinson's disease or dementia with Lewy bodies, and neuroprotective interventions are advocated. This link might also be present in patients taking antidepressants, with post-traumatic stress disorder, or with a history of repeated traumatic head injury. REM sleep likely contributes to normal memory processes. Its alteration has also been proposed to be part of the neuropathological changes occurring in Alzheimer's disease.
Collapse
Affiliation(s)
- Laura Pérez-Carbonell
- Sleep Disorders Centre, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | - Alex Iranzo
- Neurology Service, Sleep Disorders Centre, Hospital Clínic de Barcelona, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Levin J, Baiardi S, Quadalti C, Rossi M, Mammana A, Vöglein J, Bernhardt A, Perrin RJ, Jucker M, Preische O, Hofmann A, Höglinger GU, Cairns NJ, Franklin EE, Chrem P, Cruchaga C, Berman SB, Chhatwal JP, Daniels A, Day GS, Ryan NS, Goate AM, Gordon BA, Huey ED, Ibanez L, Karch CM, Lee J, Llibre‐Guerra J, Lopera F, Masters CL, Morris JC, Noble JM, Renton AE, Roh JH, Frosch MP, Keene CD, McLean C, Sanchez‐Valle R, Schofield PR, Supnet‐Bell C, Xiong C, Giese A, Hansson O, Bateman RJ, McDade E, Parchi P. α-Synuclein seed amplification assay detects Lewy body co-pathology in autosomal dominant Alzheimer's disease late in the disease course and dependent on Lewy pathology burden. Alzheimers Dement 2024; 20:4351-4365. [PMID: 38666355 PMCID: PMC11180868 DOI: 10.1002/alz.13818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Amyloid beta and tau pathology are the hallmarks of sporadic Alzheimer's disease (AD) and autosomal dominant AD (ADAD). However, Lewy body pathology (LBP) is found in ≈ 50% of AD and ADAD brains. METHODS Using an α-synuclein seed amplification assay (SAA) in cerebrospinal fluid (CSF) from asymptomatic (n = 26) and symptomatic (n = 27) ADAD mutation carriers, including 12 with known neuropathology, we investigated the timing of occurrence and prevalence of SAA positive reactivity in ADAD in vivo. RESULTS No asymptomatic participant and only 11% (3/27) of the symptomatic patients tested SAA positive. Neuropathology revealed LBP in 10/12 cases, primarily affecting the amygdala or the olfactory areas. In the latter group, only the individual with diffuse LBP reaching the neocortex showed α-synuclein seeding activity in CSF in vivo. DISCUSSION Results suggest that in ADAD LBP occurs later than AD pathology and often as amygdala- or olfactory-predominant LBP, for which CSF α-synuclein SAA has low sensitivity. HIGHLIGHTS Cerebrospinal fluid (CSF) real-time quaking-induced conversion (RT-QuIC) detects misfolded α-synuclein in ≈ 10% of symptomatic autosomal dominant Alzheimer's disease (ADAD) patients. CSF RT-QuIC does not detect α-synuclein seeding activity in asymptomatic mutation carriers. Lewy body pathology (LBP) in ADAD mainly occurs as olfactory only or amygdala-predominant variants. LBP develops late in the disease course in ADAD. CSF α-synuclein RT-QuIC has low sensitivity for focal, low-burden LBP.
Collapse
Affiliation(s)
- Johannes Levin
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Simone Baiardi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Jonathan Vöglein
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
| | - Alexander Bernhardt
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
| | - Richard J. Perrin
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Oliver Preische
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Anna Hofmann
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Günter U. Höglinger
- Department of NeurologyLMU University Hospital, LMU MunichMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Nigel J. Cairns
- Living Systems InstituteFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Erin E. Franklin
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Jasmeer P. Chhatwal
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Alisha Daniels
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Gregory S. Day
- Department of NeurologyMayo Clinic in FloridaJacksonvilleFloridaUSA
| | - Natalie S. Ryan
- Dementia Research CentreDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Alison M. Goate
- Department of Genetics & Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brian A. Gordon
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Edward D. Huey
- Butler HospitalBrown Center for Alzheimer's Disease ResearchAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Laura Ibanez
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | - Celeste M. Karch
- Department of PsychiatryWashington University School of MedicineSaint LouisMissouriUSA
| | - Jae‐Hong Lee
- Department of NeurologyAsan Medical CenterSeoulSouth Korea
| | - Jorge Llibre‐Guerra
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Francisco Lopera
- Grupo de Neurosciencias de Antioquia, Sede de Investigación Universitaria SIUMedellínColombia
| | - Colin L. Masters
- Florey Institute and The University of MelbourneMelbourneVictoriaAustralia
| | - John C. Morris
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - James M. Noble
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain, and GH Sergievsky Center, Columbia UniversityNew YorkNew YorkUSA
| | - Alan E. Renton
- Department of Genetics and Genomic Sciences and Nash Family Dept of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jee Hoon Roh
- Departments of Neurology and PhysiologyKorea University College of MedicineSeoulSouth Korea
| | - Matthew P. Frosch
- MassGeneral Institute for Neurodegenerative Diseases, Neuropathology Service, Massachusetts General HospitalBostonMassachusettsUSA
| | - C. Dirk Keene
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Catriona McLean
- Department of Anatomical PathologyAlfredHealthMelbourneVictoriaAustralia
| | - Raquel Sanchez‐Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clinic de Barcelona, FRCB‐IDIBAPSBarcelonaSpain
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Medical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Charlene Supnet‐Bell
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Chengjie Xiong
- Division of BiostatisticsWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical Sciences MalmöFaculty of Medicine, Lund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Eric McDade
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Piero Parchi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| |
Collapse
|
28
|
Biscarini F, Pizza F, Vandi S, Incensi A, Antelmi E, Donadio V, Ferri R, Liguori R, Plazzi G. Biomarkers of neurodegeneration in isolated and antidepressant-related rapid eye movement sleep behavior disorder. Eur J Neurol 2024; 31:e16260. [PMID: 38409939 PMCID: PMC11235591 DOI: 10.1111/ene.16260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/11/2024] [Accepted: 02/08/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND AND PURPOSE This study compared the features of isolated rapid eye movement (REM) sleep behavior disorder (iRBD) and antidepressant-related REM sleep behaviour disorder (RBD) with the aim of highlighting markers that might distinguish the two entities. METHODS The observational cohort study included RBD patients with and without antidepressant use (antiD+ and antiD- patients, respectively), without cognitive impairment and parkinsonism. Clinical features of RBD, subtle motor and non-motor symptoms of parkinsonism, sleep architecture, REM atonia index, dopamine transporter-single photon emission computed tomography (DAT-SPECT) and skin biopsies for the intraneuronal alpha-synuclein (α-syn), were evaluated in the baseline work-up. RESULTS Thirty-nine patients, 10 antiD+ and 29 antiD-, were included. AntiD+ patients (more frequently female) reported more psychiatric symptoms, less violent dream enactment, and less frequent hyposmia. Dermal α-syn was detected in 93.1% of antiD- versus 30% of antiD+ patients (p = 0.00024). No differences appeared in other motor and non-motor symptoms, Movement Disorder Society-Unified Parkinson's Disease Rating Scale part III score, DAT-SPECT, or polysomnographic features. CONCLUSIONS Patients with antidepressant-related RBD have clinical and neuropathological features suggesting a lower risk of evolution than those with iRBD.
Collapse
Affiliation(s)
- Francesco Biscarini
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Fabio Pizza
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Stefano Vandi
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Alex Incensi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Elena Antelmi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- DIMI Department of Engineering and Medicine of InnovationUniversity of VeronaVeronaItaly
| | - Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Raffaele Ferri
- Department of Neurology ICOasi Research Institute‐IRCCSTroinaItaly
| | - Rocco Liguori
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
| | - Giuseppe Plazzi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical, Metabolic, and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
29
|
Janarthanam C, Clabaugh G, Wang Z, Melvin BR, Scheibe I, Jin H, Anantharam V, Urbauer RJB, Urbauer JL, Ma J, Kanthasamy A, Huang X, Donadio V, Zou W, Kanthasamy AG. High-Yield α-Synuclein Purification and Ionic Strength Modification Pivotal to Seed Amplification Assay Performance and Reproducibility. Int J Mol Sci 2024; 25:5988. [PMID: 38892177 PMCID: PMC11172462 DOI: 10.3390/ijms25115988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Alpha-synuclein seed amplification assays (αSyn-SAAs) have emerged as promising diagnostic tools for Parkinson's disease (PD) by detecting misfolded αSyn and amplifying the signal through cyclic shaking and resting in vitro. Recently, our group and others have shown that multiple biospecimens, including CSF, skin, and submandibular glands (SMGs), can be used to seed the aggregation reaction and robustly distinguish between patients with PD and non-disease controls. The ultrasensitivity of the assay affords the ability to detect minute quantities of αSyn in peripheral tissues, but it also produces various technical challenges of variability. To address the problem of variability, we present a high-yield αSyn protein purification protocol for the efficient production of monomers with a low propensity for self-aggregation. We expressed wild-type αSyn in BL21 Escherichia coli, lysed the cells using osmotic shock, and isolated αSyn using acid precipitation and fast protein liquid chromatography (FPLC). Following purification, we optimized the ionic strength of the reaction buffer to distinguish the fluorescence maximum (Fmax) separation between disease and healthy control tissues for enhanced assay performance. Our protein purification protocol yielded high quantities of αSyn (average: 68.7 mg/mL per 1 L of culture) and showed highly precise and robust αSyn-SAA results using brain, skin, and SMGs with inter-lab validation.
Collapse
Affiliation(s)
- Chelva Janarthanam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Griffin Clabaugh
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Zerui Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Bradley R. Melvin
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA;
| | - Ileia Scheibe
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Huajun Jin
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Vellareddy Anantharam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Ramona J. B. Urbauer
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (R.J.B.U.); (J.L.U.)
| | - Jeffrey L. Urbauer
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (R.J.B.U.); (J.L.U.)
| | - Jiyan Ma
- Chinese Institute for Brain Research, Beijing 102206, China;
| | - Arthi Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences of Bologna, Complex Operational Unit Clinica Neurologica, 40138 Bologna, Italy;
| | - Wenquan Zou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Anumantha G. Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA; (C.J.); (G.C.); (I.S.); (H.J.); (V.A.); (A.K.)
| |
Collapse
|
30
|
Schenck CH. REM sleep behaviour disorder (RBD): Personal perspectives and research priorities. J Sleep Res 2024:e14228. [PMID: 38782758 DOI: 10.1111/jsr.14228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
The formal identification and naming of rapid eye movement (REM) sleep behaviour disorder (RBD) in 1985-1987 is described; the historical background of RBD from 1966 to 1985 is briefly discussed; and RBD milestones are presented. Current knowledge on RBD is identified with reference to recent comprehensive reviews, allowing for a focus on research priorities for RBD: factors and predictors of neurodegenerative phenoconversion from isolated RBD and patient enrolment in neuroprotective trials; isolated RBD clinical research cohorts; epidemiology of RBD; traumatic brain injury, post-traumatic stress disorder, RBD and neurodegeneration; depression, RBD and synucleinopathy; evolution of prodromal RBD to neurodegeneration; gut microbiome dysbiosis and colonic synuclein histopathology in isolated RBD; other alpha-synuclein research in isolated RBD; narcolepsy-RBD; dreams and nightmares in RBD; phasic REM sleep in isolated RBD; RBD, periodic limb movements, periodic limb movement disorder pseudo-RBD; other neurophysiology research in RBD; cardiac scintigraphy (123I-MIBG) in isolated RBD; brain magnetic resonance imaging biomarkers in isolated RBD; microRNAs as biomarkers in isolated RBD; actigraphic, other automated digital monitoring and machine learning research in RBD; prognostic counselling and ethical considerations in isolated RBD; and REM sleep basic science research. RBD research is flourishing, and is strategically situated at an ever-expanding crossroads of clinical (sleep) medicine, neurology, psychiatry and neuroscience.
Collapse
Affiliation(s)
- Carlos H Schenck
- Minnesota Regional Sleep Disorders Center, Department of Psychiatry, Hennepin County Medical Center and University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
31
|
Gibbons CH, Levine T, Adler C, Bellaire B, Wang N, Stohl J, Agarwal P, Aldridge GM, Barboi A, Evidente VGH, Galasko D, Geschwind MD, Gonzalez-Duarte A, Gil R, Gudesblatt M, Isaacson SH, Kaufmann H, Khemani P, Kumar R, Lamotte G, Liu AJ, McFarland NR, Miglis M, Reynolds A, Sahagian GA, Saint-Hillaire MH, Schwartzbard JB, Singer W, Soileau MJ, Vernino S, Yerstein O, Freeman R. Skin Biopsy Detection of Phosphorylated α-Synuclein in Patients With Synucleinopathies. JAMA 2024; 331:1298-1306. [PMID: 38506839 PMCID: PMC10955354 DOI: 10.1001/jama.2024.0792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/18/2024] [Indexed: 03/21/2024]
Abstract
Importance Finding a reliable diagnostic biomarker for the disorders collectively known as synucleinopathies (Parkinson disease [PD], dementia with Lewy bodies [DLB], multiple system atrophy [MSA], and pure autonomic failure [PAF]) is an urgent unmet need. Immunohistochemical detection of cutaneous phosphorylated α-synuclein may be a sensitive and specific clinical test for the diagnosis of synucleinopathies. Objective To evaluate the positivity rate of cutaneous α-synuclein deposition in patients with PD, DLB, MSA, and PAF. Design, Setting, and Participants This blinded, 30-site, cross-sectional study of academic and community-based neurology practices conducted from February 2021 through March 2023 included patients aged 40 to 99 years with a clinical diagnosis of PD, DLB, MSA, or PAF based on clinical consensus criteria and confirmed by an expert review panel and control participants aged 40 to 99 years with no history of examination findings or symptoms suggestive of a synucleinopathy or neurodegenerative disease. All participants completed detailed neurologic examinations and disease-specific questionnaires and underwent skin biopsy for detection of phosphorylated α-synuclein. An expert review panel blinded to pathologic data determined the final participant diagnosis. Exposure Skin biopsy for detection of phosphorylated α-synuclein. Main Outcomes Rates of detection of cutaneous α-synuclein in patients with PD, MSA, DLB, and PAF and controls without synucleinopathy. Results Of 428 enrolled participants, 343 were included in the primary analysis (mean [SD] age, 69.5 [9.1] years; 175 [51.0%] male); 223 met the consensus criteria for a synucleinopathy and 120 met criteria as controls after expert panel review. The proportions of individuals with cutaneous phosphorylated α-synuclein detected by skin biopsy were 92.7% (89 of 96) with PD, 98.2% (54 of 55) with MSA, 96.0% (48 of 50) with DLB, and 100% (22 of 22) with PAF; 3.3% (4 of 120) of controls had cutaneous phosphorylated α-synuclein detected. Conclusions and Relevance In this cross-sectional study, a high proportion of individuals meeting clinical consensus criteria for PD, DLB, MSA, and PAF had phosphorylated α-synuclein detected by skin biopsy. Further research is needed in unselected clinical populations to externally validate the findings and fully characterize the potential role of skin biopsy detection of phosphorylated α-synuclein in clinical care.
Collapse
Affiliation(s)
- Christopher H. Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Todd Levine
- HonorHealth Neurology, Scottsdale, Arizona
- CND Life Sciences, Scottsdale, Arizona
| | - Charles Adler
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, Arizona
| | | | - Ningshan Wang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | | - Georgina M. Aldridge
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Alexandru Barboi
- Department of Neurology, Northshore University Health System, Glenview, Illinois
| | | | - Douglas Galasko
- Department of Neurology, University of California, San Diego
| | | | | | - Ramon Gil
- Parkinson’s Disease Treatment Center of Southwest Florida, Port Charlotte
| | - Mark Gudesblatt
- Department of Neurology, New York University Grossman Long Island School of Medicine, New York
| | - Stuart H. Isaacson
- Parkinson’s Disease and Movement Disorders Center of Boca Raton, Boca Raton, Florida
| | - Horacio Kaufmann
- Department of Neurology, New York University Grossman School of Medicine, New York
| | - Pravin Khemani
- Department of Neurology, Swedish Medical Center, Seattle, Washington
| | - Rajeev Kumar
- Rocky Mountain Movement Disorders Center, Englewood, Colorado
| | | | - Andy J. Liu
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina
| | | | - Mitchell Miglis
- Department of Neurology, Stanford University Medical Center, Palo Alto, California
| | | | | | | | | | - Wolfgang Singer
- Department of Neurology, Mayo Clinic Rochester, Rochester, New York
| | | | - Steven Vernino
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas
| | - Oleg Yerstein
- Department of Neurology, Lahey Clinic, Burlington, Massachusetts
| | - Roy Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Painous C, Fernández M, Pérez J, de Mena L, Cámara A, Compta Y. Fluid and tissue biomarkers in Parkinson's disease: Immunodetection or seed amplification? Central or peripheral? Parkinsonism Relat Disord 2024; 121:105968. [PMID: 38168618 DOI: 10.1016/j.parkreldis.2023.105968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Over the last two decades there have been meaningful developments on biomarkers of neurodegenerative diseases, extensively (but not solely) focusing on their proteinopathic nature. Accordingly, in Alzheimer's disease determination of levels of total and phosphorylated tau (τ and p-τ, usually p-τ181) along with amyloid-beta1-42 (Aβ1-42) by immunodetection in cerebrospinal fluid (CSF) and currently even in peripheral blood, have been widely accepted and introduced to routine diagnosis. In the case of Parkinson's disease, α-synuclein as a potential biomarker (both for diagnosis and progression tracking) has proved more elusive under the immunodetection approach. In recent years, the emergence of the so-called seed amplification assays is proving to be a game-changer, with mounting evidence under different technical approaches and using a variety of biofluids or tissues, yielding promising diagnostic accuracies. Currently the least invasive but at once more reliable source of biosamples and techniques are being sought. Here we overview these advances.
Collapse
Affiliation(s)
- Celia Painous
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jesica Pérez
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Lorena de Mena
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ana Cámara
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Clínic i Universitari de Barcelona, Lab of Parkinson Disease and Other Neurodegenerative Movement Disorders - Clinical and Experimental Research, IDIBAPS, Institut de Neurociències UBNeuro, Universitat de Barcelona, Barcelona, Catalonia, Spain.
| |
Collapse
|
33
|
Kulcsarova K, Skorvanek M. Challenges and Future of the International Parkinson and Movement Disorder Society Prodromal Parkinson's Disease Criteria: Are We On the Right Track? Mov Disord 2024; 39:637-643. [PMID: 38310367 DOI: 10.1002/mds.29724] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/05/2024] Open
Affiliation(s)
- Kristina Kulcsarova
- Department of Neurology, P. J. Safarik University, Kosice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Kosice, Slovakia
- Department of Clinical Neurosciences, University Scientific Park MEDIPARK, P. J. Safarik University, Kosice, Slovakia
| | - Matej Skorvanek
- Department of Neurology, P. J. Safarik University, Kosice, Slovakia
- Department of Neurology, University Hospital of L. Pasteur, Kosice, Slovakia
| |
Collapse
|
34
|
Wang Z, Wu L, Gerasimenko M, Gilliland T, Gunzler SA, Donadio V, Liguori R, Xu B, Zou WQ. Seeding Activity of Skin Misfolded Tau as a Biomarker for Tauopathies. RESEARCH SQUARE 2024:rs.3.rs-3968879. [PMID: 38496453 PMCID: PMC10942562 DOI: 10.21203/rs.3.rs-3968879/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Background Tauopathies are a group of age-related neurodegenerative diseases characterized by the accumulation of pathologically phosphorylated tau protein in the brain, leading to prion-like propagation and aggregation. They include Alzheimer's disease (AD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD). Currently, reliable diagnostic biomarkers that directly reflect the capability of propagation and spreading of misfolded tau aggregates in peripheral tissues and body fluids are lacking. Methods We utilized the seed-amplification assay (SAA) employing ultrasensitive real-time quaking-induced conversion (RT-QuIC) to assess the prion-like seeding activity of pathological tau in the skin of cadavers with neuropathologically confirmed tauopathies, including AD, PSP, CBD, and PiD, compared to normal controls. Results We found that the skin prion-SAA demonstrated a significantly higher sensitivity (75-80%) and specificity (95-100%) for detecting tauopathy, depending on the tau substrates used. Moreover, increased tau-seeding activity was also observed in biopsy skin samples from living AD and PSP patients examined. Analysis of the end products of skin-tau SAA confirmed that the increased seeding activity was accompanied by the formation of tau aggregates with different physicochemical properties related to two different tau substrates used. Conclusions Overall, our study provides proof-of-concept that the skin tau-SAA can differentiate tauopathies from normal controls, suggesting that the seeding activity of misfolded tau in the skin could serve as a diagnostic biomarker for tauopathies.
Collapse
Affiliation(s)
- Zerui Wang
- Case Western Reserve University School of Medicine
| | - Ling Wu
- North Carolina Central University
| | | | | | - Steven A Gunzler
- University Hospitals Cleveland Medical Center: UH Cleveland Medical Center
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences of Bolgna: IRCCS Istituto Delle Scienze Neurologiche di Bologna
| | - Rocco Liguori
- IRCCS Institute of Neurological Sciences of Bologna: IRCCS Istituto Delle Scienze Neurologiche di Bologna
| | - Bin Xu
- North Carolina Central University
| | - Wen-Quan Zou
- First Affiliated Hospital of Nanchang University
| |
Collapse
|
35
|
Tóth Š, Kulcsárová K, Maretta M, Kunová A, Mechírová E, Gdovinová Z, Feketeová E, Ribeiro Ventosa J, Baloghová J, Bekeová M, Christová P, Mrázová S, Muránska S, Zeidan D, Škorvánek M. α-synuclein antibody 5G4 identifies idiopathic REM-sleep behavior disorder in abdominal skin biopsies. Parkinsonism Relat Disord 2024; 120:105956. [PMID: 38217955 DOI: 10.1016/j.parkreldis.2023.105956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Idiopathic REM-sleep behavior disorder (iRBD) is considered the most specific prodromal marker of Parkinson's disease (PD). With the need to improve early detection of prodromal α-synucleinopathies, several methods to identify peripheral α-synuclein (α-syn) pathology have been exploited in manifest and prodromal PD with varying diagnostic accuracy. Recently, a disease specific 5G4 antibody has been evaluated in skin biopsies of manifest PD patients. The aim of our study was to analyze the 5G4 α-syn immunoreactivity in skin biopsies of deeply phenotyped subjects with iRBD and controls. METHODS The study cohort consisted of 28 patients with PD, 24 subjects with iRBD and 27 healthy controls, recruited from the CEGEMOD, PDBIOM and PARCAS cohorts. All subjects were deeply phenotyped and assessed for prodromal PD (pPD) probability based on MDS research criteria. Abdominal skin punch biopsies were processed and stained using a conformation specific 5G4 α-syn antibody as well as axonal markers SMI-31 and S100. RESULTS 5G4-positivity was identified in 23/28 PD patients, 20/24 iRBD subjects and 8/27 healthy controls. Compared to healthy controls, sensitivity and specificity reached 83.33 % and 70.37 % for iRBD; and 82.14 % and 70.37 % for PD, respectively. 5G4-positivity rate in our study was irrespective of the calculated pPD probability of iRBD subjects. CONCLUSIONS This work establishes the diagnostic yield of conformation specific 5G4 α-syn antibody testing in skin biopsies of subjects with pPD, specifically iRBD. The diagnostic accuracy for this method seems to be similar for both manifest and prodromal PD and is not dependent on the pPD probability ratios.
Collapse
Affiliation(s)
- Štefan Tóth
- Department of Histology and Embryology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic.
| | - Kristína Kulcsárová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic; Department of Clinical Neurosciences, University Scientific Park MEDIPARK, P. J. Šafárik University, Kosice, Slovak Republic
| | - Milan Maretta
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic
| | - Alexandra Kunová
- Department of Histology and Embryology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Eva Mechírová
- Department of Histology and Embryology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Zuzana Gdovinová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic
| | - Eva Feketeová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic
| | - Joaquim Ribeiro Ventosa
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic; Department of Clinical Neurosciences, University Scientific Park MEDIPARK, P. J. Šafárik University, Kosice, Slovak Republic
| | - Janette Baloghová
- Department of Dermatovenerology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Dermatovenerology, University Hospital of L. Pasteur, Košice, Slovak Republic
| | - Martina Bekeová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Petronela Christová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Soňa Mrázová
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Soňa Muránska
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Dema Zeidan
- Department of Histology and Embryology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic
| | - Matej Škorvánek
- Department of Neurology, Medical Faculty of P. J. Šafárik University, Košice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Košice, Slovak Republic
| |
Collapse
|
36
|
Huang J, Yuan X, Chen L, Hu B, Wang H, Wang Y, Huang W. Pathological α-synuclein detected by real-time quaking-induced conversion in synucleinopathies. Exp Gerontol 2024; 187:112366. [PMID: 38280659 DOI: 10.1016/j.exger.2024.112366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/10/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
synucleinopathies are diseases characterized by the aggregation of α-synuclein (α-syn), which forms fibrils through misfolding and accumulates in a prion-like manner. To detect the presence of these α-syn aggregates in clinical samples, seed amplification assays (SAAs) have been developed. These SAAs are capable of amplifying the α-syn seeds, allowing for their detection. αSyn-SAAs have been reported under the names 'protein misfolding cyclic amplification' (αSyn-PMCA) and 'real-time quaking-induced conversion'α-Syn-RT-QuIC. The α-Syn RT-QuIC, in particular, has been adapted to amplify and detect α-syn aggregates in various biospecimens, including cerebrospinal fluid (CSF), skin, nasal brushing, serum and saliva. The α-syn RT-QuIC assay has demonstrated good sensitivity and specificity in detecting pathological α-syn, particularly in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) cases, with an accuracy rate of up to 80 %. Additionally, differential diagnosis between DLB and PD, as well as PD and multiple system atrophy (MSA), can be achieved by utilizing certain kinetic thioflavin T (ThT) parameters and other parameters. Moreover, the positive detection of α-syn in the prodromal stage of synucleinopathies provides an opportunity for early intervention and management. In summary, the development of the α-syn RT-QuIC assay has greatly contributed to the field of synucleinopathies. Therefore, we review the development of α-syn RT-QuIC assay and describe in detail the recent advancements of α-syn RT-QuIC assay for detecting pathological α-syn in synucleinopathies.
Collapse
Affiliation(s)
- Juan Huang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Xingxing Yuan
- Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, China
| | - Lin Chen
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Binbin Hu
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Hui Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China
| | - Ye Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China.
| | - Wei Huang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
37
|
Brockmann K, Lerche S, Baiardi S, Rossi M, Wurster I, Quadalti C, Roeben B, Mammana A, Zimmermann M, Hauser AK, Deuschle C, Schulte C, Liepelt-Scarfone I, Gasser T, Parchi P. CSF α-synuclein seed amplification kinetic profiles are associated with cognitive decline in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:24. [PMID: 38242875 PMCID: PMC10799016 DOI: 10.1038/s41531-023-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024] Open
Abstract
Seed amplification assays have been implemented in Parkinson's disease to reveal disease-specific misfolded alpha-synuclein aggregates in biospecimens. While the assays' qualitative dichotomous seeding response is valuable to stratify and enrich cohorts for alpha-synuclein pathology in general, more quantitative parameters that are associated with clinical dynamics of disease progression and that might potentially serve as exploratory outcome measures in clinical trials targeting alpha-synuclein would add important information. To evaluate whether the seeding kinetic parameters time required to reach the seeding threshold (LAG phase), the peak of fluorescence response (Imax), and the area under the curve (AUC) are associated with clinical trajectories, we analyzed LAG, Imax, and AUC in relation to the development of cognitive decline in a longitudinal cohort of 199 people with Parkinson's disease with positive CSF alpha-synuclein seeding status. Patients were stratified into tertiles based on their individual CSF alpha-synuclein seeding kinetic properties. The effect of the kinetic parameters on longitudinal development of cognitive impairment defined by MoCA ≤25 was analyzed by Cox-Regression. Patients with a higher number of positive seeding replicates and tertile groups of shorter LAG, higher Imax, and higher AUC showed a higher prevalence of and a shorter duration until cognitive impairment longitudinally (3, 6, and 3 years earlier with p ≤ 0.001, respectively). Results remained similar in separate subgroup analyses of patients with and without GBA mutation. We conclude that a more prominent alpha-synuclein seeding kinetic profile translates into a more rapid development of cognitive decline.
Collapse
Affiliation(s)
- Kathrin Brockmann
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany.
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany.
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
| | - Stefanie Lerche
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
| | - Isabel Wurster
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
- Edmond J. Safra Fellow in Movement Disorders, Tuebingen, Germany
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Benjamin Roeben
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
| | - Milan Zimmermann
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Ann-Kathrin Hauser
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Christian Deuschle
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Claudia Schulte
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Inga Liepelt-Scarfone
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Thomas Gasser
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, University of Tuebingen, Hoppe Seyler‑Strasse 3, 72076, Tuebingen, Germany
- German Center for Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 1/8, 40139, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
| |
Collapse
|
38
|
Bentivenga GM, Mammana A, Baiardi S, Rossi M, Ticca A, Magliocchetti F, Mastrangelo A, Poleggi A, Ladogana A, Capellari S, Parchi P. Performance of a seed amplification assay for misfolded alpha-synuclein in cerebrospinal fluid and brain tissue in relation to Lewy body disease stage and pathology burden. Acta Neuropathol 2024; 147:18. [PMID: 38240849 PMCID: PMC10799141 DOI: 10.1007/s00401-023-02663-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 01/22/2024]
Abstract
The development of in vitro seed amplification assays (SAA) detecting misfolded alpha-synuclein (αSyn) in cerebrospinal fluid (CSF) and other tissues has provided a pathology-specific biomarker for Lewy body disease (LBD). However, αSyn SAA diagnostic performance in early pathological stages or low Lewy body (LB) pathology load has only been assessed in small cohorts. Moreover, the relationship between SAA kinetic parameters, the number of αSyn brain seeds and the LB pathology burden assessed by immunohistochemistry has never been systematically investigated. We tested 269 antemortem CSF samples and 138 serially diluted brain homogenates from patients with and without neuropathological evidence of LBD in different stages by the αSyn Real-Time Quaking-Induced Conversion (RT-QuIC) SAA. Moreover, we looked for LB pathology by αSyn immunohistochemistry in a consecutive series of 604 Creutzfeldt-Jakob disease (CJD)-affected brains. αSyn CSF RT-QuIC showed 100% sensitivity in detecting LBD in limbic and neocortical stages. The assay sensitivity was significantly lower in patients in early stages (37.5% in Braak 1 and 2, 73.3% in Braak 3) or with focal pathology (50% in amygdala-predominant). The average number of CSF RT-QuIC positive replicates significantly correlated with LBD stage. Brain homogenate RT-QuIC showed higher sensitivity than immunohistochemistry for the detection of misfolded αSyn. In the latter, the kinetic parameter lag phase (time to reach the positive threshold) strongly correlated with the αSyn seed concentration in serial dilution experiments. Finally, incidental LBD prevalence was 8% in the CJD cohort. The present results indicate that (a) CSF RT-QuIC has high specificity and sufficient sensitivity to detect all patients with LB pathology at Braak stages > 3 and most of those at stage 3; (b) brain deposition of misfolded αSyn precedes the formation of LB and Lewy neurites; (c) αSyn SAA provides "quantitative" information regarding the LB pathology burden, with the lag phase and the number of positive replicates being the most promising variables to be used in the clinical setting.
Collapse
Affiliation(s)
| | - Angela Mammana
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marcello Rossi
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | - Andrea Mastrangelo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Poleggi
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Ladogana
- Department of Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Sabina Capellari
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Piero Parchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
- IRCCS, Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy.
| |
Collapse
|
39
|
Yan S, Jiang C, Janzen A, Barber TR, Seger A, Sommerauer M, Davis JJ, Marek K, Hu MT, Oertel WH, Tofaris GK. Neuronally Derived Extracellular Vesicle α-Synuclein as a Serum Biomarker for Individuals at Risk of Developing Parkinson Disease. JAMA Neurol 2024; 81:59-68. [PMID: 38048087 PMCID: PMC10696516 DOI: 10.1001/jamaneurol.2023.4398] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/28/2023] [Indexed: 12/05/2023]
Abstract
IMPORTANCE Nonmotor symptoms of Parkinson disease (PD) often predate the movement disorder by decades. Currently, there is no blood biomarker to define this prodromal phase. OBJECTIVE To investigate whether α-synuclein in neuronally derived serum-extracellular vesicles identifies individuals at risk of developing PD and related dementia. DESIGN, SETTING, and PARTICIPANTS This retrospective, cross-sectional multicenter study of serum samples included the Oxford Discovery, Marburg, Cologne, and Parkinson's Progression Markers Initiative cohorts. Participants were recruited from July 2013 through August 2023 and samples were analyzed from April 2022 through September 2023. The derivation group (n = 170) included participants with isolated rapid eye movement sleep behavior disorder (iRBD) and controls. Two validation groups were used: the first (n = 122) included participants with iRBD and controls and the second (n = 263) included nonmanifest GBA1N409S gene carriers, participants with iRBD or hyposmia, and available dopamine transporter single-photon emission computed tomography, healthy controls, and patients with sporadic PD. Overall the study included 199 participants with iRBD, 20 hyposmic participants with available dopamine transporter single-photon emission computed tomography, 146 nonmanifest GBA1N409S gene carriers, 21 GBA1N409S gene carrier patients with PD, 50 patients with sporadic PD, and 140 healthy controls. In the derivation group and validation group 1, participants with polysomnographically confirmed iRBD were included. In the validation group 2, at-risk participants with available Movement Disorder Society prodromal markers and serum samples were included. Among 580 potential participants, 4 were excluded due to alternative diagnoses. EXPOSURES Clinical assessments, imaging, and serum collection. MAIN OUTCOME AND MEASURES L1CAM-positive extracellular vesicles (L1EV) were immunocaptured from serum. α-Synuclein and syntenin-1 were measured by electrochemiluminescence. Area under the receiver operating characteristic (ROC) curve (AUC) with 95% CIs evaluated biomarker performance. Probable prodromal PD was determined using the updated Movement Disorder Society research criteria. Multiple linear regression models assessed the association between L1EV α-synuclein and prodromal markers. RESULTS Among 576 participants included, the mean (SD) age was 64.30 (8.27) years, 394 were male (68.4%), and 182 were female (31.6%). A derived threshold of serum L1EV α-synuclein distinguished participants with iRBD from controls (AUC = 0.91; 95% CI, 0.86-0.96) and those with more than 80% probability of having prodromal PD from participants with less than 5% probability (AUC = 0.80; 95% CI, 0.71-0.89). Subgroup analyses revealed that specific combinations of prodromal markers were associated with increased L1EV α-synuclein levels. Across all cohorts, L1EV α-synuclein differentiated participants with more than 80% probability of having prodromal PD from current and historic healthy control populations (AUC = 0.90; 95% CI, 0.87-0.93), irrespective of initial diagnosis. L1EV α-synuclein was increased in at-risk participants with a positive cerebrospinal fluid seed amplification assay and was above the identified threshold in 80% of cases (n = 40) that phenoconverted to PD or related dementia. CONCLUSIONS AND RELEVANCE L1EV α-synuclein in combination with prodromal markers should be considered in the stratification of those at high risk of developing PD and related Lewy body diseases.
Collapse
Affiliation(s)
- Shijun Yan
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| | - Cheng Jiang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| | - Annette Janzen
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Thomas R. Barber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| | - Aline Seger
- Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany
| | - Michael Sommerauer
- Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany
| | - Jason J. Davis
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Michele T. Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| | | | - George K. Tofaris
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Pilotto A, Zanusso G, Antelmi E, Okuzumi A, Zatti C, Lupini A, Bongianni M, Padovani A, Hattori N. Biofluid Markers and Tissue Biopsies Analyses for the Prodromal and Earliest Phase of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S333-S344. [PMID: 39331105 PMCID: PMC11494635 DOI: 10.3233/jpd-240007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/28/2024]
Abstract
The recent development of new methods to detect misfolded α-synuclein (αSyn) aggregates in biofluids and tissue biopsies in the earliest Parkinson's disease (PD) phases is dramatically challenging the biological definition of PD. The αSyn seed amplification methods in cerebrospinal fluid (CSF) showed high sensitivity and specificity for early diagnosis of PD and Lewy bodies disorders. Several studies in isolated REM sleep behavior disorders and other at-risk populations also demonstrated a high prevalence of CSF αSyn positivity and its potential value in predicting the phenoconversion to clinically manifested diseases. Growing evidence exists for αSyn aggregates in olfactory mucosa, skin, and other tissues in subjects with PD or at-risk subjects. DOPA decarboxylase and numerous other candidates have been additionally proposed for either diagnostic or prognostic purposes in earliest PD phases. The newly described αSyn detection in blood, through its quantification in neuronally-derived exosome vesicles, represents a technical challenge that could open a new scenario for the biological diagnosis of PD. Despite this growing evidence in the field, most of method of αSyn detection and markers still need to be validated in ongoing longitudinal studies through an accurate assessment of different prodromal disease subtypes and scenarios before being definitively implemented in clinical settings.
Collapse
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gianluigi Zanusso
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Elena Antelmi
- Neurology Unit, Parkinson Disease and Movement Disorders Division, Department of Engineering and Medicine of Innovation, University of Verona, Verona, Italy
| | - Ayami Okuzumi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Cinzia Zatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Alessandro Lupini
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Matilde Bongianni
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Neurobiorepository and Laboratory of Advanced Biological Markers, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Neurodegenerative Disorders Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
41
|
Kluge A, Iranzo A. Biofluid Detection of Pathological α-Synuclein in the Prodromal Phase of Synucleinopathies. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S323-S331. [PMID: 38995801 PMCID: PMC11494638 DOI: 10.3233/jpd-230429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 07/14/2024]
Abstract
Synucleinopathies are disorders characterized by the aggregation and deposition of pathological α-synuclein conformers. The underlying neurodegenerative processes begin years or decades before the onset of cardinal motor symptoms. This prodromal phase may manifest with various signs or symptoms. However, there are no current standardized laboratory tests to ascertain the progression and conversion of prodromal conditions such as mild cognitive impairment, isolated REM sleep behavior disorder or pure autonomic failure. The aim of this systematic review was to evaluate the diagnostic possibilities using human biofluids as source material to detect pathological α-synuclein in the prodromal phase of synucleinopathies. Our review identified eight eligible studies, that investigated pathological α-synuclein conformers using cerebrospinal fluid from patients with prodromal signs of synulceinopathies to differentiate this patient group from non-synucleinopathies, while only one study investigated this aspect using blood as medium. While previous studies clearly demonstrated a high diagnostic performance of α-synuclein seed amplification assays for differentiating synucleinopathies with Lewy bodies from healthy controls, only few analyses were performed focussing on individuals with prodromal disease. Nevertheless, results for the early detection of α-synuclein seeds using α-synuclein seed amplification assays were promising and may be of particular relevance for future clinical trials and clinical practice.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Kiel University, Kiel, Germany
| | - Alex Iranzo
- Sleep Unit, Neurology Service, Hospital Clínic Barcelona, Barcelona University, IDIBAPS, CIBERNED, Barcelona, Spain
| |
Collapse
|
42
|
Schaeffer E, Yilmaz R, St. Louis EK, Noyce AJ. Ethical Considerations for Identifying Individuals in the Prodromal/Early Phase of Parkinson's Disease: A Narrative Review. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S307-S319. [PMID: 38995800 PMCID: PMC11492008 DOI: 10.3233/jpd-230428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
The ability to identify individuals in the prodromal phase of Parkinson's disease has improved in recent years, raising the question of whether and how those affected should be informed about the risk of future disease. Several studies investigated prognostic counselling for individuals with isolated REM sleep behavior disorder and have shown that most patients want to receive information about prognosis, but autonomy and individual preferences must be respected. However, there are still many unanswered questions about risk disclosure or early diagnosis of PD, including the impact on personal circumstances, cultural preferences and specific challenges associated with different profiles of prodromal symptoms, genetic testing or biomarker assessments. This narrative review aims to summarize the current literature on prognostic counselling and risk disclosure in PD, as well as highlight future perspectives that may emerge with the development of new biomarkers and their anticipated impact on the definition of PD.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Kiel University, Kiel, Germany
| | - Rezzak Yilmaz
- Department of Neurology, Ankara University School of Medicine, Ankara, Turkey
- Ankara University Brain Research Center, Ankara, Turkey
| | - Erik K. St. Louis
- Mayo Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, USA
| | - Alastair J. Noyce
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
43
|
Orso B, Brosse S, Frasnelli J, Arnaldi D. Opportunities and Pitfalls of REM Sleep Behavior Disorder and Olfactory Dysfunction as Early Markers in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S275-S285. [PMID: 38517805 PMCID: PMC11494648 DOI: 10.3233/jpd-230348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 03/24/2024]
Abstract
During its pre-motor stage, Parkinson's disease (PD) presents itself with a multitude of non-motor symptoms with different degrees of specificity and sensitivity. The most important among them are REM sleep behavior disorder (RBD) and olfactory dysfunction. RBD is a parasomnia characterized by the loss of REM sleep muscle atonia and dream-enacting behaviors. Olfactory dysfunction in individuals with prodromal PD is usually described as hyposmia (reduced sense of smell) or anosmia (complete loss of olfactory function). These symptoms can precede the full expression of motor symptoms by decades. A close comprehension of these symptoms and the underlying mechanisms may enable early screening as well as interventions to improve patients' quality of life. Therefore, these symptoms have unmatched potential for identifying PD patients in prodromal stages, not only allowing early diagnosis but potentially opening a window for early, possibly disease-modifying intervention. However, they come with certain challenges. This review addresses some of the key opportunities and pitfalls of both RBD and olfactory dysfunction as early markers of PD.
Collapse
Affiliation(s)
- Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Clinical Neurology, University of Genoa, Genoa, Italy
| | - Sarah Brosse
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
- Research Center, Sacré-Coeur Hospital of Montreal, Montréal, Québec, Canada
| | - Johannes Frasnelli
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
- Research Center, Sacré-Coeur Hospital of Montreal, Montréal, Québec, Canada
| | - Dario Arnaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Clinical Neurology, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| |
Collapse
|
44
|
Weintraub D. What's in a Name? The Time Has Come to Unify Parkinson's Disease and Dementia with Lewy Bodies. Mov Disord 2023; 38:1977-1981. [PMID: 37614069 DOI: 10.1002/mds.29590] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Affiliation(s)
- Daniel Weintraub
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
45
|
Roig-Uribe M, Serradell M, Muñoz-Lopetegi A, Gaig C, Iranzo A. Prior exposure to concussions in patients with isolated REM sleep behavior disorder. Sleep Med 2023; 110:254-257. [PMID: 37660513 DOI: 10.1016/j.sleep.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/18/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE Traumatic brain injury is associated with the late development of neurodegenerative diseases such as the synucleinopathies. Isolated REM sleep behavior disorder (IRBD) constitutes an early manifestation of the synucleinopathies. We assessed whether lifetime history of concussive episodes is common in IRBD and examined its characteristics and clinical significance. METHODS Prior exposure to concussions was evaluated by interviewing polysomnographically-confirmed IRBD patients and controls without IRBD, and by the BRAIN-Q questionnaire. RESULTS We recruited 199 IRBD patients aged 73.2 ± 7.7 years and 168 age and sex matched controls. Previous history of concussion was more common in patients than in controls (21.1% versus 10.1%, p = 0.004). In patients, concussions occurred at the age of 24.7 ± 20.6 years. The interval between concussion and IRBD diagnosis was 43.0 ± 19.0 years. There were no differences between patients and controls in the causes of concussions (e.g., traffic accidents, sport practice), and number of events resulting in skull fractures, urgent medical assistance, and hospitalization. After a follow-up of 5.7 ± 4.7 years from IRBD diagnosis, 21.1% patients developed an overt synucleinopathy with an interval of 49.3 ± 24.2 years between concussion and synucleinopathy diagnosis. The risk to develop a synucleinopathy was similar between patients with and without concussions (p = 0.57). CONCLUSIONS Previous history of concussion is common in IRBD. Our observations may suggest that in individuals with increased susceptibility, early-life concussions may trigger a slow neurodegenerative process leading four decades later to IRBD. This study highlights the need for head injury prevention, particularly in early life.
Collapse
Affiliation(s)
- Mónica Roig-Uribe
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain
| | - Mònica Serradell
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain
| | - Amaia Muñoz-Lopetegi
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain
| | - Carles Gaig
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain
| | - Alex Iranzo
- Sleep Disorders Center, Neurology Service, Hospital Clínic Barcelona, Universitat de Barcelona, IDIBAPS, CIBERNED: CB06/05/0018-ISCIII, Barcelona, Spain.
| |
Collapse
|
46
|
Schließer P, Struebing FL, Northoff BH, Kurz A, Rémi J, Holdt L, Höglinger GU, Herms J, Koeglsperger T. Detection of a Parkinson's Disease-Specific MicroRNA Signature in Nasal and Oral Swabs. Mov Disord 2023; 38:1706-1715. [PMID: 37382573 DOI: 10.1002/mds.29515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/11/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Biomaterials from oral and nasal swabs provide, in theory, a potential resource for biomarker development. However, their diagnostic value has not yet been investigated in the context of Parkinson's disease (PD) and associated conditions. OBJECTIVE We have previously identified a PD-specific microRNA (miRNA) signature in gut biopsies. In this work, we aimed to investigate the expression of miRNAs in routine buccal (oral) and nasal swabs obtained from cases with idiopathic PD and isolated rapid eye movement sleep behavior disorder (iRBD), a prodromal symptom that often precedes α-synucleinopathies. We aimed to address their value as a diagnostic biomarker for PD and their mechanistic contribution to PD onset and progression. METHODS Healthy control cases (n = 28), cases with PD (n = 29), and cases with iRBD (n = 8) were prospectively recruited to undergo routine buccal and nasal swabs. Total RNA was extracted from the swab material, and the expression of a predefined set of miRNAs was quantified by quantitative real-time polymerase chain reaction. RESULTS Statistical analysis revealed a significantly increased expression of hsa-miR-1260a in cases who had PD. Interestingly, hsa-miR-1260a expression levels correlated with diseases severity, as well as olfactory function, in the PD and iRBD cohorts. Mechanistically, hsa-miR-1260a segregated to Golgi-associated cellular processes with a potential role in mucosal plasma cells. Predicted hsa-miR-1260a target gene expression was reduced in iRBD and PD groups. CONCLUSIONS Our work demonstrates oral and nasal swabs as a valuable biomarker pool in PD and associated neurodegenerative conditions. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patricia Schließer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Felix L Struebing
- Department of Translational Brain Research, German Centre for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig Maximilian University, Munich, Germany
| | - Bernd H Northoff
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Kurz
- Department of Gynaecology and Obstetrics, Klinikum Landsberg am Lech, Landsberg, Germany
| | - Jan Rémi
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lesca Holdt
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases e.V. (DZNE) Munich, Munich, Germany
| | - Jochen Herms
- Department of Translational Brain Research, German Centre for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig Maximilian University, Munich, Germany
| | - Thomas Koeglsperger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Translational Brain Research, German Centre for Neurodegenerative Diseases, Munich, Germany
| |
Collapse
|
47
|
Tsalenchuk M, Gentleman SM, Marzi SJ. Linking environmental risk factors with epigenetic mechanisms in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:123. [PMID: 37626097 PMCID: PMC10457362 DOI: 10.1038/s41531-023-00568-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Sporadic Parkinson's disease (PD) is a progressive neurodegenerative disease, with a complex risk structure thought to be influenced by interactions between genetic variants and environmental exposures, although the full aetiology is unknown. Environmental factors, including pesticides, have been reported to increase the risk of developing the disease. Growing evidence suggests epigenetic changes are key mechanisms by which these environmental factors act upon gene regulation, in disease-relevant cell types. We present a systematic review critically appraising and summarising the current body of evidence of the relationship between epigenetic mechanisms and environmental risk factors in PD to inform future research in this area. Epigenetic studies of relevant environmental risk factors in animal and cell models have yielded promising results, however, research in humans is just emerging. While published studies in humans are currently relatively limited, the importance of the field for the elucidation of molecular mechanisms of pathogenesis opens clear and promising avenues for the future of PD research. Carefully designed epidemiological studies carried out in PD patients hold great potential to uncover disease-relevant gene regulatory mechanisms. Therefore, to advance this burgeoning field, we recommend broadening the scope of investigations to include more environmental exposures, increasing sample sizes, focusing on disease-relevant cell types, and recruiting more diverse cohorts.
Collapse
Affiliation(s)
- Maria Tsalenchuk
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK.
- Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
48
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Santos-García D, Martínez-Valbuena I, Agúndez JAG. Alpha-Synuclein in Peripheral Tissues as a Possible Marker for Neurological Diseases and Other Medical Conditions. Biomolecules 2023; 13:1263. [PMID: 37627328 PMCID: PMC10452242 DOI: 10.3390/biom13081263] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The possible usefulness of alpha-synuclein (aSyn) determinations in peripheral tissues (blood cells, salivary gland biopsies, olfactory mucosa, digestive tract, skin) and in biological fluids, except for cerebrospinal fluid (serum, plasma, saliva, feces, urine), as a marker of several diseases, has been the subject of numerous publications. This narrative review summarizes data from studies trying to determine the role of total, oligomeric, and phosphorylated aSyn determinations as a marker of various diseases, especially PD and other alpha-synucleinopathies. In summary, the results of studies addressing the determinations of aSyn in its different forms in peripheral tissues (especially in platelets, skin, and digestive tract, but also salivary glands and olfactory mucosa), in combination with other potential biomarkers, could be a useful tool to discriminate PD from controls and from other causes of parkinsonisms, including synucleinopathies.
Collapse
Affiliation(s)
| | | | - Elena García-Martín
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - Diego Santos-García
- Department of Neurology, CHUAC—Complejo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain;
| | - Iván Martínez-Valbuena
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 2S8, Canada;
| | - José A. G. Agúndez
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
49
|
Quadalti C, Palmqvist S, Hall S, Rossi M, Mammana A, Janelidze S, Dellavalle S, Mattsson-Carlgren N, Baiardi S, Stomrud E, Hansson O, Parchi P. Clinical effects of Lewy body pathology in cognitively impaired individuals. Nat Med 2023; 29:1964-1970. [PMID: 37464058 PMCID: PMC10427416 DOI: 10.1038/s41591-023-02449-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
There is poor knowledge about the clinical effects of Lewy body (LB) pathology in patients with cognitive impairment, especially when coexisting with Alzheimer's disease (AD) pathology (amyloid-β and tau). Using a seed amplification assay, we analyzed cerebrospinal fluid for misfolded LB-associated α-synuclein in 883 memory clinic patients with mild cognitive impairment or dementia from the BioFINDER study. Twenty-three percent had LB pathology, of which only 21% fulfilled clinical criteria of Parkinson's disease or dementia with Lewy bodies at baseline. Among these LB-positive patients, 48% had AD pathology. Fifty-four percent had AD pathology in the whole sample (17% of mild cognitive impairment and 24% of patients with dementia were also LB-positive). When examining independent cross-sectional effects, LB pathology but not amyloid-β or tau, was associated with hallucinations and worse attention/executive, visuospatial and motor function. LB pathology was also associated with faster longitudinal decline in all examined cognitive functions, independent of amyloid-β, tau, cognitive stage and a baseline diagnosis of dementia with Lewy bodies/Parkinson's disease. LB status provides a better precision-medicine approach to predict clinical trajectories independent of AD biomarkers and a clinical diagnosis, which could have implications for the clinical management of cognitive impairment and the design of AD and LB drug trials.
Collapse
Affiliation(s)
- Corinne Quadalti
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Sofia Dellavalle
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Neurology Clinic, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Simone Baiardi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
50
|
Coughlin DG, Irwin DJ. Fluid and Biopsy Based Biomarkers in Parkinson's Disease. Neurotherapeutics 2023; 20:932-954. [PMID: 37138160 PMCID: PMC10457253 DOI: 10.1007/s13311-023-01379-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Several advances in fluid and tissue-based biomarkers for use in Parkinson's disease (PD) and other synucleinopathies have been made in the last several years. While work continues on species of alpha-synuclein (aSyn) and other proteins which can be measured from spinal fluid and plasma samples, immunohistochemistry and immunofluorescence from peripheral tissue biopsies and alpha-synuclein seeding amplification assays (aSyn-SAA: including real-time quaking induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA)) now offer a crucial advancement in their ability to identify aSyn species in PD patients in a categorical fashion (i.e., of aSyn + vs aSyn -); to augment clinical diagnosis however, aSyn-specific assays that have quantitative relevance to pathological burden remain an unmet need. Alzheimer's disease (AD) co-pathology is commonly found postmortem in PD, especially in those who develop dementia, and dementia with Lewy bodies (DLB). Biofluid biomarkers for tau and amyloid beta species can detect AD co-pathology in PD and DLB, which does have relevance for prognosis, but further work is needed to understand the interplay of aSyn tau, amyloid beta, and other pathological changes to generate comprehensive biomarker profiles for patients in a manner translatable to clinical trial design and individualized therapies.
Collapse
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, 9444 Medical Center Drive, ECOB 03-021, MCC 0886, La Jolla, CA, 92037, USA.
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|