1
|
Jiang LL, Zhang XL, Hu HY. Co-Aggregation of TDP-43 with Other Pathogenic Proteins and Their Co-Pathologies in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:12380. [PMID: 39596445 PMCID: PMC11594478 DOI: 10.3390/ijms252212380] [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: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Pathological aggregation of a specific protein into insoluble aggregates is a common hallmark of various neurodegenerative diseases (NDDs). In the earlier literature, each NDD is characterized by the aggregation of one or two pathogenic proteins, which can serve as disease-specific biomarkers. The aggregation of these specific proteins is thought to be a major cause of or deleterious result in most NDDs. However, accumulating evidence shows that a pathogenic protein can interact and co-aggregate with other pathogenic proteins in different NDDs, thereby contributing to disease onset and progression synergistically. During the past years, more than one type of NDD has been found to co-exist in some individuals, which may increase the complexity and pathogenicity of these diseases. This article reviews and discusses the biochemical characteristics and molecular mechanisms underlying the co-aggregation and co-pathologies associated with TDP-43 pathology. The TDP-43 aggregates, as a hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), can often be detected in other NDDs, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and spinocerebellar ataxia type 2 (SCA2). In many cases, TDP-43 is shown to interact and co-aggregate with multiple pathogenic proteins in vitro and in vivo. Furthermore, the co-occurrence and co-aggregation of TDP-43 with other pathogenic proteins have important consequences that may aggravate the diseases. Thus, the current viewpoint that the co-aggregation of TDP-43 with other pathogenic proteins in NDDs and their relevance to disease progression may gain insights into the patho-mechanisms and therapeutic potential of various NDDs.
Collapse
Affiliation(s)
- Lei-Lei Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| | - Xiang-Le Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-Yu Hu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| |
Collapse
|
2
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [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: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
3
|
Anastassiadis C, Martinez-Valbuena I, Vasilevskaya A, Thapa S, Hadian M, Morales-Rivero A, Mora-Fisher D, Salvo C, Taghdiri F, Sato C, Moreno D, Anor CJ, Misquitta K, Couto B, Tang-Wai DF, Lang AE, Fox SH, Rogaeva E, Kovacs GG, Tartaglia MC. CSF α-Synuclein Seed Amplification Assay in Patients With Atypical Parkinsonian Disorders. Neurology 2024; 103:e209818. [PMID: 39208367 DOI: 10.1212/wnl.0000000000209818] [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: 09/04/2024] Open
Abstract
BACKGROUND AND OBJECTIVES There is no disease-modifying treatment of corticobasal syndrome (CBS) and progressive supranuclear palsy (PSP), 2 disorders characterized by their striking phenotypic, and, in CBS, pathologic heterogeneity. Seed amplification assays (SAAs) could enable the detection of neuropathologic processes, such as α-synuclein (αSyn) copathology, that affect the success of future disease-modifying treatment strategies. The primary objective was to assess possible αSyn copathology in CBS and PSP, as detected in CSF using an αSyn SAA (αSyn-SAA). Secondary objectives were to evaluate the association of αSyn-SAA positivity with other biomarkers including of Alzheimer disease (AD), and with clinical presentation. We hypothesized that αSyn-SAA positivity would be detectable in CBS and PSP and that it would be associated with AD biomarker positivity and β-amyloid (Aβ) 42 levels, neurodegeneration as assessed by neurofilament light chain (NfL) levels, and symptoms associated with synucleinopathies. METHODS This cross-sectional observational study included patients clinically diagnosed with CBS and PSP who underwent a lumbar puncture between 2012 and 2021 (Toronto Western Hospital, Canada). CSF was tested for αSyn-SAA positivity, AD biomarkers, and NfL levels. Clinical data were derived from medical records. RESULTS We tested the CSF of 40 patients with CBS (19 female patients, 65.9 ± 8.6 years) and 28 with PSP (13 female patients, 72.5 ± 8.7 years old), mostly White (n = 50) or Asian (n = 14). αSyn-SAA positivity was observed in 35.9% patients with CBS and 28.6% with PSP. In young-onset, but not late-onset patients, αSyn-SAA positivity and AD positivity were associated (odds ratio [OR] 8.8, 95% CI 1.2-82.6, p < 0.05). A multivariable linear regression analysis showed a significant interaction of αSyn-SAA status by age at onset on CSF Aβ42 levels (β = 0.3 ± 0.1, p < 0.05). Indeed, age at onset was positively related to Aβ42 levels only in αSyn-SAA-positive patients, as shown by slope comparison. A logistic regression analysis also suggested that REM sleep behavior disorder was associated with αSyn-SAA positivity (OR 60.2, 95% CI 5.2-1,965.8; p < 0.01). DISCUSSION We detected a frequency of αSyn-SAA positivity in CBS and PSP in line with pathologic studies, highlighting the usefulness of SAAs for in vivo detection of otherwise undetectable neuropathologic processes. Our results also suggest that AD status (specifically low Aβ42) and older age at onset may contribute to αSyn-SAA positivity. This opens new perspectives for the stratification of patients in clinical trials.
Collapse
Affiliation(s)
- Chloe Anastassiadis
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Ivan Martinez-Valbuena
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Anna Vasilevskaya
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Simrika Thapa
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Mohsen Hadian
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Alonso Morales-Rivero
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Daniela Mora-Fisher
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Cristina Salvo
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Foad Taghdiri
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Christine Sato
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Danielle Moreno
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Cassandra J Anor
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Karen Misquitta
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Blas Couto
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - David F Tang-Wai
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Anthony E Lang
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Susan H Fox
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Ekaterina Rogaeva
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Gabor G Kovacs
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- From the Tanz Centre for Research in Neurodegenerative Diseases (C.A., I.M.-V., A.V., S.T., M.H., F.T., C. Sato, D.M., C.J.A., K.M., A.E.L., E.R., G.G.K., M.C.T.); Krembil Brain Institute (I.M.-V., A.M.-R., B.C., D.F.T.-W., A.E.L., S.H.F., G.G.K., M.C.T.); The Edmond J. Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders Clinic (I.M.-V., A.M.-R., B.C., A.E.L., S.H.F., G.G.K., M.C.T.); Rossy Progressive Supranuclear Palsy Centre (I.M.-V., A.M.-R., A.E.L., G.G.K., M.C.T.), University Health Network and the University of Toronto; and University Health Network Memory Clinic (D.M.-F., C. Salvo, D.F.T.-W.), Toronto, Ontario, Canada
| |
Collapse
|
4
|
Nishitani T, Mitoh Y, Yajima T, Tachiya D, Hoshika T, Fukunaga T, Nishitani Y, Yoshida R, Mizoguchi I, Ichikawa H, Sato T. Distribution of alpha-synuclein in rat salivary glands. Anat Rec (Hoboken) 2024; 307:2933-2946. [PMID: 38284507 DOI: 10.1002/ar.25395] [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: 10/25/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Expression of alpha-synuclein (Syn), a presynaptic neuronal protein, was immunohistochemically examined in intact rat submandibular, sublingual, and lingual glands. The submandibular gland contained abundant periductal Syn-immunoreactive (-ir) nerve fibers. Abundant Syn-ir varicosities were present in acini of the sublingual and serous lingual glands. By confocal laser scanning microscopy, Syn-ir nerve fibers around smooth muscle actin (SMA)-ir cells alone were infrequent; however, those around aquaporin-5 (AQP5)-ir cells alone and both SMA- and AQP5-ir cells were abundant in the sublingual and serous lingual glands. SMA-ir cells were occasionally immunoreactive for toll-like receptor 4, a Syn receptor. Syn-ir nerve fibers contained tyrosine hydroxylase (TH) in the submandibular gland and choline acetyltransferase (ChAT) in all examined salivary glands. In the superior cervical (SCG), submandibular, and intralingual ganglia, sympathetic and parasympathetic neurons co-expressed Syn with TH and ChAT, respectively. SCG neurons innervating the submandibular gland contained mostly Syn. In the thoracic spinal cord, 14.7% of ChAT-ir preganglionic sympathetic neurons co-expressed Syn. In the superior salivatory nucleus, preganglionic parasympathetic neurons projecting to the lingual nerve co-expressed Syn and ChAT. The present findings indicate that released Syn acts on myoepithelial cells. Syn in pre- and post-ganglionic neurons may regulate neurotransmitter release and salivary volume and composition.
Collapse
Affiliation(s)
- Tomiko Nishitani
- Divisions of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai, Japan
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Yoshihiro Mitoh
- Department of Oral Physiology, Okayama University Graduate School of Medicine and Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takehiro Yajima
- Divisions of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Daisuke Tachiya
- Divisions of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Tomohiro Hoshika
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Tomohiro Fukunaga
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Okayama University Graduate School of Medicine and Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Itaru Mizoguchi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Hiroyuki Ichikawa
- Divisions of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Tadasu Sato
- Divisions of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| |
Collapse
|
5
|
Kobylecki C, Thompson JC, Robinson AC, Roncaroli F, Snowden JS, Mann DM. Concomitant progressive supranuclear palsy and Lewy body pathology presenting with circumscribed visual memory loss: A clinicopathological case. Brain Pathol 2024; 34:e13219. [PMID: 37927160 PMCID: PMC11189767 DOI: 10.1111/bpa.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
A 70-year-old man presented to the clinic with impairment of visual memory and marked predominantly right sided mesial temporal lobe atrophy on imaging. He died 6 years following symptom onset and neuropathological examination showed concomitant progressive supranuclear palsy and Lewy body pathology. Although he did not fulfil clinical criteria for either condition at presentation, we propose that interactions between the two pathologies in mesial temporal regions could result in this atypical clinical phenotype.
Collapse
Affiliation(s)
- Christopher Kobylecki
- Department of Neurology, Manchester Centre for Clinical NeurosciencesNorthern Care Alliance NHS Foundation TrustSalfordUK
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Jennifer C. Thompson
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Cerebral Function Unit, Manchester Centre for Clinical NeurosciencesNorthern Care Alliance NHS Foundation TrustSalfordUK
| | - Andrew C. Robinson
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Federico Roncaroli
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Neuropathology Unit, Manchester Centre for Clinical NeurosciencesNorthern Care Alliance NHS Foundation TrustSalfordUK
| | - Julie S. Snowden
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Cerebral Function Unit, Manchester Centre for Clinical NeurosciencesNorthern Care Alliance NHS Foundation TrustSalfordUK
| | - David M. Mann
- Division of Neuroscience, Geoffrey Jefferson Brain Research Centre, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
6
|
Schreiber CS, Wiesweg I, Stanelle-Bertram S, Beck S, Kouassi NM, Schaumburg B, Gabriel G, Richter F, Käufer C. Sex-specific biphasic alpha-synuclein response and alterations of interneurons in a COVID-19 hamster model. EBioMedicine 2024; 105:105191. [PMID: 38865747 PMCID: PMC11293593 DOI: 10.1016/j.ebiom.2024.105191] [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: 12/13/2023] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) frequently leads to neurological complications after recovery from acute infection, with higher prevalence in women. However, mechanisms by which SARS-CoV-2 disrupts brain function remain unclear and treatment strategies are lacking. We previously demonstrated neuroinflammation in the olfactory bulb of intranasally infected hamsters, followed by alpha-synuclein and tau accumulation in cortex, thus mirroring pathogenesis of neurodegenerative diseases such as Parkinson's or Alzheimer's disease. METHODS To uncover the sex-specific spatiotemporal profiles of neuroinflammation and neuronal dysfunction following intranasal SARS-CoV-2 infection, we quantified microglia cell density, alpha-synuclein immunoreactivity and inhibitory interneurons in cortical regions, limbic system and basal ganglia at acute and late post-recovery time points. FINDINGS Unexpectedly, microglia cell density and alpha-synuclein immunoreactivity decreased at 6 days post-infection, then rebounded to overt accumulation at 21 days post-infection. This biphasic response was most pronounced in amygdala and striatum, regions affected early in Parkinson's disease. Several brain regions showed altered densities of parvalbumin and calretinin interneurons which are involved in cognition and motor control. Of note, females appeared more affected. INTERPRETATION Our results demonstrate that SARS-CoV-2 profoundly disrupts brain homeostasis without neuroinvasion, via neuroinflammatory and protein regulation mechanisms that persist beyond viral clearance. The regional patterns and sex differences are in line with neurological deficits observed after SARS-CoV-2 infection. FUNDING Federal Ministry of Health, Germany (BMG; ZMV I 1-2520COR501 to G.G.), Federal Ministry of Education and Research, Germany (BMBF; 03COV06B to G.G.), Ministry of Science and Culture of Lower Saxony in Germany (14-76403-184, to G.G. and F.R.).
Collapse
Affiliation(s)
- Cara Sophie Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany
| | - Ivo Wiesweg
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Sebastian Beck
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Nancy Mounogou Kouassi
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Berfin Schaumburg
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Gülsah Gabriel
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany; Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| |
Collapse
|
7
|
Monchi O, Pinilla-Monsalve GD, Almgren H, Ghahremani M, Kibreab M, Maarouf N, Kathol I, Boré A, Rheault F, Descoteaux M, Ismail Z. White Matter Microstructural Underpinnings of Mild Behavioral Impairment in Parkinson's Disease. Mov Disord 2024; 39:1026-1036. [PMID: 38661496 DOI: 10.1002/mds.29804] [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: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Patients with Parkinson's disease (PD) experience changes in behavior, personality, and cognition that can manifest even in the initial stages of the disease. Previous studies have suggested that mild behavioral impairment (MBI) should be considered an early marker of cognitive decline. However, the precise neurostructural underpinnings of MBI in early- to mid-stage PD remain poorly understood. OBJECTIVE The aim was to explore the changes in white matter microstructure linked to MBI and mild cognitive impairment (MCI) in early- to mid-stage PD using diffusion magnetic resonance imaging (dMRI). METHODS A total of 91 PD patients and 36 healthy participants were recruited and underwent anatomical MRI and dMRI, a comprehensive neuropsychological battery, and the completion of the Mild Behavioral Impairment-Checklist. Metrics of white matter integrity included tissue fractional anisotropy (FAt) and radial diffusivity (RDt), free water (FW), and fixel-based apparent fiber density (AFD). RESULTS The connection between the left amygdala and the putamen was disrupted when comparing PD patients with MBI (PD-MBI) to PD-non-MBI, as evidenced by increased RDt (η2 = 0.09, P = 0.004) and both decreased AFD (η2 = 0.05, P = 0.048) and FAt (η2 = 0.12, P = 0.014). Compared to controls, PD patients with both MBI and MCI demonstrated increased FW for the connection between the left orbitofrontal gyrus (OrG) and the hippocampus (η2 = 0.22, P = 0.008), augmented RDt between the right OrG and the amygdala (η2 = 0.14, P = 0.008), and increased RDt (η2 = 0.25, P = 0.028) with decreased AFD (η2 = 0.10, P = 0.046) between the right OrG and the caudate nucleus. CONCLUSION MBI is associated with abnormal microstructure of connections involving the orbitofrontal cortex, putamen, and amygdala. To our knowledge, this is the first assessment of the white matter microstructure in PD-MBI using dMRI. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Oury Monchi
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada
- Département de radiologie, radio-oncologie et médicine nucléaire, Université de Montréal, Montreal, Quebec, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Gabriel D Pinilla-Monsalve
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada
- Département de radiologie, radio-oncologie et médicine nucléaire, Université de Montréal, Montreal, Quebec, Canada
| | - Hannes Almgren
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Maryam Ghahremani
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Psychiatry and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Mekale Kibreab
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Nadia Maarouf
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Iris Kathol
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Arnaud Boré
- Département d'informatique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - François Rheault
- Département d'informatique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Maxime Descoteaux
- Département d'informatique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Zahinoor Ismail
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Psychiatry and Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
8
|
Venkatesh Y, Marotta NP, Lee VMY, Petersson EJ. Highly tunable bimane-based fluorescent probes: design, synthesis, and application as a selective amyloid binding dye. Chem Sci 2024; 15:6053-6063. [PMID: 38665526 PMCID: PMC11040648 DOI: 10.1039/d4sc00024b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
Small molecule fluorescent probes are indispensable tools for a broad range of biological applications. Despite many probes being available, there is still a need for probes where photophysical properties and biological selectivity can be tuned as desired. Here, we report the rational design and synthesis of a palette of fluorescent probes based on the underexplored bimane scaffold. The newly developed probes with varied electronic properties show tunable absorption and emission in the visible region with large Stokes shifts. Probes featuring electron-donating groups exhibit rotor effects that are sensitive to polarity and viscosity by "intramolecular charge transfer" (ICT) and twisted intramolecular charge transfer (TICT) mechanisms, respectively. These properties enable their application as "turn-on" fluorescent probes to detect fibrillar aggregates of the α-synuclein (αS) protein that are a hallmark of Parkinson's disease (PD). One probe shows selective binding to αS fibrils relative to soluble proteins in cell lysates and amyloid fibrils of tau and amyloid-β. Finally, we demonstrate the diagnostic potential of the probe in selectively detecting αS fibrils amplified from PD with dementia (PDD) patient samples.
Collapse
Affiliation(s)
- Yarra Venkatesh
- Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104 USA
| | - Nicholas P Marotta
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania 3600 Spruce Street Philadelphia PA 19104 USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania 3600 Spruce Street Philadelphia PA 19104 USA
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104 USA
- Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania 421 Curie Boulevard Philadelphia PA 19104 USA
| |
Collapse
|
9
|
Aliakbari F, Marzookian K, Parsafar S, Hourfar H, Nayeri Z, Fattahi A, Raeiji M, Boroujeni NN, Otzen DE, Morshedi D. The impact of hUC MSC-derived exosome-nanoliposome hybrids on α-synuclein fibrillation and neurotoxicity. SCIENCE ADVANCES 2024; 10:eadl3406. [PMID: 38569030 PMCID: PMC10990263 DOI: 10.1126/sciadv.adl3406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
Amyloid aggregation of α-synuclein (αSN) protein amplifies the pathogenesis of neurodegenerative diseases (NDs) such as Parkinson's disease (PD). Consequently, blocking aggregation or redirecting self-assembly to less toxic aggregates could be therapeutic. Here, we improve brain-specific nanocarriers using a hybrid of exosomes (Ex) from human umbilical cord mesenchymal stem cells (hUC MSCs) and nanoliposomes containing baicalein (Ex-NLP-Ba) and oleuropein (Ex-NLP-Ole). The hybrids contained both lipid membranes, Ex proteins, and baicalein or oleuropein. Fluorescence resonance energy transfer analysis confirmed their proper integration. The hybrids reduced the extent of αSN fibrillation and interfered with secondary nucleation and disaggregation. They not only reduced αSN pathogenicity but also enhanced drug internalization into cells, surpassing the efficacy of NLP alone, and also crossed the blood-brain barrier in a cellular model. We conclude that Ex can be successfully extracted and efficiently merged with NLPs while retaining its original properties, demonstrating great potential as a theranostic drug delivery vehicle against NDs like PD.
Collapse
Affiliation(s)
- Farhang Aliakbari
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Kimia Marzookian
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Soha Parsafar
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hamdam Hourfar
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Nayeri
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Arghavan Fattahi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad Raeiji
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Narges Nasrollahi Boroujeni
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Dina Morshedi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
10
|
Melachroinou K, Divolis G, Tsafaras G, Karampetsou M, Fortis S, Stratoulias Y, Papadopoulou G, Kriebardis AG, Samiotaki M, Vekrellis K. Endogenous Alpha-Synuclein is Essential for the Transfer of Pathology by Exosome-Enriched Extracellular Vesicles, Following Inoculation with Preformed Fibrils in vivo. Aging Dis 2024; 15:869-892. [PMID: 37548944 PMCID: PMC10917543 DOI: 10.14336/ad.2023.0614] [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: 04/07/2023] [Accepted: 06/14/2023] [Indexed: 08/08/2023] Open
Abstract
The main pathological hallmark of Parkinson's disease (PD) and related synucleinopathies is the presence of intracellular proteinaceous aggregates, enriched in the presynaptic protein alpha-Synuclein (α-Syn). α-Syn association with exosomes has been previously documented both as a physiological process of secretion and as a pathological process of disease transmission, however, critical information about the mechanisms governing this interplay is still lacking. To address this, we utilized the α-Syn preformed fibril (PFF) mouse model of PD, as a source of brain-derived exosome-enriched extracellular vesicles (ExE-EVs) and assessed their pathogenic capacity following intrastriatal injections in host wild type (WT) mouse brain. We further investigated the impact of the fibrillar α-Syn on the exosomal cargo independent of the endogenous α-Syn, by isolating ExE-EVs from PFF-injected α-Syn knockout mice. Although PFF inoculation does not alter the morphology, size distribution, and quantity of brain-derived ExE-EVs, it triggers changes in the exosomal proteome related to synaptic and mitochondrial function, as well as metabolic processes. Importantly, we showed that the presence of the endogenous α-Syn is essential for the ExE-EVs to acquire a pathogenic capacity, allowing them to mediate disease transmission by inducing phosphorylated-α-Syn pathology. Notably, misfolded α-Syn containing ExE-EVs when injected in WT mice were able to induce astrogliosis and synaptic alterations in the host brain, at very early stages of α-Syn pathology, preceding the formation of the insoluble α-Syn accumulations. Collectively, our data suggest that exosomal cargo defines their ability to spread α-Syn pathology.
Collapse
Affiliation(s)
- Katerina Melachroinou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Georgios Divolis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - George Tsafaras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Mantia Karampetsou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Sotirios Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece.
| | - Yannis Stratoulias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Gina Papadopoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Kostas Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
11
|
Smith R, Hovren H, Bowser R, Bakkar N, Garruto R, Ludolph A, Ravits J, Gaertner L, Murphy D, Lebovitz R. Misfolded alpha-synuclein in amyotrophic lateral sclerosis: Implications for diagnosis and treatment. Eur J Neurol 2024; 31:e16206. [PMID: 38270442 PMCID: PMC11235862 DOI: 10.1111/ene.16206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/30/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Alpha-synuclein (α-Syn) oligomers and fibrils have been shown to augment the aggregation of TAR DNA-binding Protein 43 (TDP-43) monomers in vitro, supporting the idea that TDP-43 proteinopathies such as ALS may be modulated by the presence of toxic forms of α-Syn. Recently, parkinsonian features were reported in a study of European patients and Lewy bodies have been demonstrated pathologically in a similar series of patients. Based on these and other considerations, we sought to determine whether seed-competent α-Syn can be identified in spinal fluid of patients with ALS including familial, sporadic, and Guamanian forms of the disease. METHODS Based on the finding that α-Syn has been found to be a prion-like protein, we have utilized a validated α-Synuclein seed amplification assay to determine if seed-competent α-Syn could be detected in the spinal fluid of patients with ALS. RESULTS Toxic species of α-Syn were detected in CSF in 18 of 127 ALS patients, 5 of whom were from Guam. Two out of twenty six samples from patients with C9orf72 variant ALS had positive seed-amplification assays (SAAs). No positive tests were noted in superoxide dismutase type 1 ALS subjects (n = 14). The SAA was negative in 31 control subjects. CONCLUSIONS Our findings suggest that a sub-group of ALS occurs in which self-replicating α-Syn is detectable and likely contributes to its pathogenesis. This finding may have implications for the diagnosis and treatment of this disorder.
Collapse
Affiliation(s)
| | - Hanna Hovren
- Amprion Clinical LaboratorySan DiegoCaliforniaUSA
| | | | | | | | | | - John Ravits
- University of California, San DiegoLa JollaCaliforniaUSA
| | - Lia Gaertner
- Bay Area Lyme Disease FoundationPortola ValleyCaliforniaUSA
| | - Davan Murphy
- Center for Neurologic StudyLa JollaCaliforniaUSA
| | | |
Collapse
|
12
|
Adam H, Gopinath SCB, Arshad MKM, Adam T, Subramaniam S, Hashim U. An Update on Parkinson's Disease and its Neurodegenerative Counterparts. Curr Med Chem 2024; 31:2770-2787. [PMID: 37016529 DOI: 10.2174/0929867330666230403085733] [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: 11/23/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Neurodegenerative disorders are a group of diseases that cause nerve cell degeneration in the brain, resulting in a variety of symptoms and are not treatable with drugs. Parkinson's disease (PD), prion disease, motor neuron disease (MND), Huntington's disease (HD), spinal cerebral dyskinesia (SCA), spinal muscle atrophy (SMA), multiple system atrophy, Alzheimer's disease (AD), spinocerebellar ataxia (SCA) (ALS), pantothenate kinase-related neurodegeneration, and TDP-43 protein disorder are examples of neurodegenerative diseases. Dementia is caused by the loss of brain and spinal cord nerve cells in neurodegenerative diseases. BACKGROUND Even though environmental and genetic predispositions have also been involved in the process, redox metal abuse plays a crucial role in neurodegeneration since the preponderance of symptoms originates from abnormal metal metabolism. METHOD Hence, this review investigates several neurodegenerative diseases that may occur symptoms similar to Parkinson's disease to understand the differences and similarities between Parkinson's disease and other neurodegenerative disorders based on reviewing previously published papers. RESULTS Based on the findings, the aggregation of alpha-synuclein occurs in Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. Other neurodegenerative diseases occur with different protein aggregation or mutations. CONCLUSION We can conclude that Parkinson's disease, Multiple system atrophy, and Dementia with Lewy bodies are closely related. Therefore, researchers must distinguish among the three diseases to avoid misdiagnosis of Multiple System Atrophy and Dementia with Lewy bodies with Parkinson's disease symptoms.
Collapse
Affiliation(s)
- Hussaini Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
| | - Subash C B Gopinath
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600, Arau, Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600, Arau, Perlis, Malaysia
- Centre for Chemical Biology (CCB), Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia
| | - M K Md Arshad
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia
| | - Tijjani Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600, Arau, Perlis, Malaysia
| | - Sreeramanan Subramaniam
- School of Biological Sciences, Universiti Sains Malaysia, Georgetown, 11800 Penang, Malaysia
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600, Arau, Perlis, Malaysia
- Centre for Chemical Biology (CCB), Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia
- National Poison Centre, Universiti Sains Malaysia (USM), Georgetown, 11800, Penang, Malaysia
| | - Uda Hashim
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
| |
Collapse
|
13
|
Alexoudi A, Kesidou L, Gatzonis S, Charalampopoulos C, Tsoga A. Effectiveness of the Combination of Probiotic Supplementation on Motor Symptoms and Constipation in Parkinson's Disease. Cureus 2023; 15:e49320. [PMID: 38146566 PMCID: PMC10749423 DOI: 10.7759/cureus.49320] [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] [Accepted: 11/23/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) reflects the second most common neurodegenerative disorder and is associated with high morbidity and mortality. Besides the motor features, non-motor symptoms, such as constipation, are very common. There is accumulating evidence that neuroinflammation is associated with the PD pathological processes. Alterations of gut microbiota and microbial metabolites have been linked to the pathogenesis of PD. Previous research has shown that probiotic supplementation has beneficial effects on motor and non-motor symptoms and especially on constipation. AIM In this study, we examine the effectiveness of a combination of probiotic supplementation (butyrate triglyceride 302.86 mg, Crocus sativus L. 30 mg, and vitamin D3 100 mcg), on constipation and motor symptoms in PD. METHODS The present study is a retrospective study that examined the existing medical records of patients with diagnosed PD, having chronic constipation and used the probiotic supplementation for its management. A total of 41 existing medical records were screened. Medical records were excluded in the case of participation in another study for PD, suffering from irritable bowel syndrome, organic constipation, long-term laxative use changes in the standard dopaminergic treatment, Mini-Mental Status Examination (MMSE) score<24, hospitalization and antibiotic medication, and diarrheal syndrome. Nine medical records were excluded, and a final number of 32 medical records was finally examined. All 32 patients had evaluations carried out at baseline and three months after supplement administration. A stool diary questionnaire, the Unified Parkinson's Disease Rating Scale III (UPDRS III), and the Schwab and England and the Hoehn and Yahr scales were used for the evaluation. RESULTS The median defecation frequency was significantly improved. The supplementation administration significantly improved UPDRS III by 7.7% (from 35.72±15.51 to 32.97±15.71, p = 0.005) at month three, as compared to baseline. A positive effect was also seen in the Schwab and England scale. There was no effect on the Hoehn and Yahr scale. CONCLUSION The enteric microbiome composition is altered in PD, and there is accumulating evidence that probiotic supplementation could alleviate disease symptoms in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Athanasia Alexoudi
- Department of Neurosurgery, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, GRC
- Neurology, Neurological Institute of Athens (NIA), Athens, GRC
| | - Lydia Kesidou
- Dentistry, Santorini General Hospital, Santorini, GRC
| | - Stylianos Gatzonis
- Department of Neurosurgery, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, GRC
| | | | - Areti Tsoga
- Department of Public Health Policy, University of West Attica, Athens, GRC
| |
Collapse
|
14
|
Leitão ADG, Ahammad RU, Spencer B, Wu C, Masliah E, Rissman RA. Novel systemic delivery of a peptide-conjugated antisense oligonucleotide to reduce α-synuclein in a mouse model of Alzheimer's disease. Neurobiol Dis 2023; 186:106285. [PMID: 37690676 PMCID: PMC10584037 DOI: 10.1016/j.nbd.2023.106285] [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: 04/18/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
Neurodegenerative disorders of aging are characterized by the progressive accumulation of proteins such as α-synuclein (α-syn) and amyloid beta (Aβ). Misfolded and aggregated α-syn has been implicated in neurological disorders such as Parkinson's disease, and Dementia with Lewy Bodies, but less so in Alzheimer's Disease (AD), despite the fact that accumulation of α-syn has been confirmed in over 50% of postmortem brains neuropathologically diagnosed with AD. To date, no therapeutic strategy has effectively or consistently downregulated α-syn in AD. Here we tested the hypothesis that by using a systemically-delivered peptide (ApoB11) bound to a modified antisense oligonucleotide against α-syn (ASO-α-syn), we can downregulate α-syn expression in an AD mouse model and improve behavioral and neuropathologic phenotypes. Our results demonstrate that monthly systemic treatment with of ApoB11:ASO α-syn beginning at 6 months of age reduces expression of α-synuclein in the brains of 9-month-old AD mice. Downregulation of α-syn led to reduction in Aβ plaque burden, prevented neuronal loss and astrogliosis. Furthermore, we found that AD mice treated with ApoB11:ASO α-syn had greatly improved hippocampal and spatial memory function in comparison to their control counterparts. Collectively, our data supports the reduction of α-syn through use of systemically-delivered ApoB11:ASO α-syn as a promising future disease-modifying therapeutic for AD.
Collapse
Affiliation(s)
- André D G Leitão
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Rijwan U Ahammad
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, United States of America
| | - Eliezer Masliah
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD 20892, United States of America
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, United States of America; VA San Diego Healthcare System, San Diego, CA 92161, United States of America.
| |
Collapse
|
15
|
Graves NJ, Gambin Y, Sierecki E. α-Synuclein Strains and Their Relevance to Parkinson's Disease, Multiple System Atrophy, and Dementia with Lewy Bodies. Int J Mol Sci 2023; 24:12134. [PMID: 37569510 PMCID: PMC10418915 DOI: 10.3390/ijms241512134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Like many neurodegenerative diseases, Parkinson's disease (PD) is characterized by the formation of proteinaceous aggregates in brain cells. In PD, those proteinaceous aggregates are formed by the α-synuclein (αSyn) and are considered the trademark of this neurodegenerative disease. In addition to PD, αSyn pathological aggregation is also detected in atypical Parkinsonism, including Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA), as well as neurodegeneration with brain iron accumulation, some cases of traumatic brain injuries, and variants of Alzheimer's disease. Collectively, these (and other) disorders are referred to as synucleinopathies, highlighting the relation between disease type and protein misfolding/aggregation. Despite these pathological relationships, however, synucleinopathies cover a wide range of pathologies, present with a multiplicity of symptoms, and arise from dysfunctions in different neuroanatomical regions and cell populations. Strikingly, αSyn deposition occurs in different types of cells, with oligodendrocytes being mainly affected in MSA, while aggregates are found in neurons in PD. If multiple factors contribute to the development of a pathology, especially in the cases of slow-developing neurodegenerative disorders, the common presence of αSyn aggregation, as both a marker and potential driver of disease, is puzzling. In this review, we will focus on comparing PD, DLB, and MSA, from symptomatology to molecular description, highlighting the role and contribution of αSyn aggregates in each disorder. We will particularly present recent evidence for the involvement of conformational strains of αSyn aggregates and discuss the reciprocal relationship between αSyn strains and the cellular milieu. Moreover, we will highlight the need for effective methodologies for the strainotyping of aggregates to ameliorate diagnosing capabilities and therapeutic treatments.
Collapse
Affiliation(s)
| | | | - Emma Sierecki
- EMBL Australia Node for Single Molecule Sciences and School of Biomedical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW 2052, Australia; (N.J.G.)
| |
Collapse
|
16
|
Zhang T, Luu MDA, Dolga AM, Eisel ULM, Schmidt M. The old second messenger cAMP teams up with novel cell death mechanisms: potential translational therapeutical benefit for Alzheimer's disease and Parkinson's disease. Front Physiol 2023; 14:1207280. [PMID: 37405135 PMCID: PMC10315612 DOI: 10.3389/fphys.2023.1207280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent neurodegenerative disorders severely impacting life expectancy and quality of life of millions of people worldwide. AD and PD exhibit both a very distinct pathophysiological disease pattern. Intriguingly, recent researches, however, implicate that overlapping mechanisms may underlie AD and PD. In AD and PD, novel cell death mechanisms, encompassing parthanatos, netosis, lysosome-dependent cell death, senescence and ferroptosis, apparently rely on the production of reactive oxygen species, and seem to be modulated by the well-known, "old" second messenger cAMP. Signaling of cAMP via PKA and Epac promotes parthanatos and induces lysosomal cell death, while signaling of cAMP via PKA inhibits netosis and cellular senescence. Additionally, PKA protects against ferroptosis, whereas Epac1 promotes ferroptosis. Here we review the most recent insights into the overlapping mechanisms between AD and PD, with a special focus on cAMP signaling and the pharmacology of cAMP signaling pathways.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Minh D. A. Luu
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ulrich L. M. Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
17
|
Di Leva F, Filosi M, Oyston L, Silvestri E, Picard A, Lavdas AA, Lobbestael E, Baekelandt V, Neely GG, Pramstaller PP, Hicks AA, Corti C. Increased Levels of the Parkinson's Disease-Associated Gene ITPKB Correlate with Higher Expression Levels of α-Synuclein, Independent of Mutation Status. Int J Mol Sci 2023; 24:1984. [PMID: 36768321 PMCID: PMC9916293 DOI: 10.3390/ijms24031984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
Autosomal dominant mutations in the gene encoding α-synuclein (SNCA) were the first to be linked with hereditary Parkinson's disease (PD). Duplication and triplication of SNCA has been observed in PD patients, together with mutations at the N-terminal of the protein, among which A30P and A53T influence the formation of fibrils. By overexpressing human α-synuclein in the neuronal system of Drosophila, we functionally validated the ability of IP3K2, an ortholog of the GWAS identified risk gene, Inositol-trisphosphate 3-kinase B (ITPKB), to modulate α-synuclein toxicity in vivo. ITPKB mRNA and protein levels were also increased in SK-N-SH cells overexpressing wild-type α-synuclein, A53T or A30P mutants. Kinase overexpression was detected in the cytoplasmatic and in the nuclear compartments in all α-synuclein cell types. By quantifying mRNAs in the cortex of PD patients, we observed higher levels of ITPKB mRNA when SNCA was expressed more (p < 0.05), compared to controls. A positive correlation was also observed between SNCA and ITPKB expression in the cortex of patients, which was not seen in the controls. We replicated this observation in a public dataset. Our data, generated in SK-N-SH cells and in cortex from PD patients, show that the expression of α-synuclein and ITPKB is correlated in pathological situations.
Collapse
Affiliation(s)
- Francesca Di Leva
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Michele Filosi
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Lisa Oyston
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Erica Silvestri
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Anne Picard
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Alexandros A. Lavdas
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - G. Gregory Neely
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Peter P. Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Andrew A. Hicks
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| | - Corrado Corti
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, 39100 Bolzano, Italy
| |
Collapse
|
18
|
Herman S, Djaldetti R, Mollenhauer B, Offen D. CSF-derived extracellular vesicles from patients with Parkinson's disease induce symptoms and pathology. Brain 2023; 146:209-224. [PMID: 35881523 DOI: 10.1093/brain/awac261] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease is characterized by the gradual appearance of intraneuronal inclusions that are primarily composed of misfolded α-synuclein protein, leading to cytotoxicity and neural death. Recent in vitro and in vivo studies suggest that misfolded α-synuclein may spread transcellularly in a prion-like manner, inducing pathological aggregates in healthy neurons, and is disseminated via secretion of extracellular vesicles. Accordingly, extracellular vesicles derived from brain lysates and CSF of patients with Parkinson's disease were shown to facilitate α-synuclein aggregation in healthy cells. Prompted by the hypothesis of Braak and colleagues that the olfactory bulb is one of the primary propagation sites for the initiation of Parkinson's disease, we sought to investigate the role of extracellular vesicles in the spread of α-synuclein and progression of Parkinson's disease through the olfactory bulb. Extracellular vesicles derived from the CSF of patients diagnosed with Parkinson's disease or with a non-synucleinopathy neurodegenerative disorder were administered intranasally to healthy mice, once daily over 4 days. Three months later, mice were subjected to motor and non-motor tests. Functional impairments were elucidated by histochemical analysis of midbrain structures relevant to Parkinson's disease pathology, 8 months after EVs treatment. Mice treated with extracellular vesicles from the patients with Parkinson's disease displayed multiple symptoms consistent with prodromal and clinical-phase Parkinson's disease such as hyposmia, motor behaviour impairments and high anxiety levels. Furthermore, their midbrains showed widespread α-synuclein aggregations, dopaminergic neurodegeneration, neuroinflammation and altered autophagy activity. Several unconventional pathologies were also observed, such as α-synuclein aggregations in the red nucleus, growth of premature grey hair and astrogliosis. Collectively, these data indicate that intranasally administered extracellular vesicles derived from the CSF of patients with Parkinson's disease can propagate α-synuclein aggregation in vivo and trigger Parkinson's disease-like symptoms and pathology in healthy mice.
Collapse
Affiliation(s)
- Shay Herman
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ruth Djaldetti
- Department of Neurology, Rabin Medical Center-Beilinson Hospital, Petach Tikva 4941492, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.,Paracelsus-Elena-Klinik, Kassel, Germany
| | - Daniel Offen
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, and Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv 6997801, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
19
|
Mazzetti S, Calogero AM, Pezzoli G, Cappelletti G. Cross-talk between α-synuclein and the microtubule cytoskeleton in neurodegeneration. Exp Neurol 2023; 359:114251. [PMID: 36243059 DOI: 10.1016/j.expneurol.2022.114251] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 12/30/2022]
Abstract
Looking at the puzzle that depicts the molecular determinants in neurodegeneration, many pieces are lacking and multiple interconnections among key proteins and intracellular pathways still remain unclear. Here we focus on the concerted action of α-synuclein and the microtubule cytoskeleton, whose interplay, indeed, is emerging but remains largely unexplored in both its physiology and pathology. α-Synuclein is a key protein involved in neurodegeneration, underlying those diseases termed synucleinopathies. Its propensity to interact with other proteins and structures renders the identification of neuronal death trigger extremely difficult. Conversely, the unbalance of microtubule cytoskeleton in terms of structure, dynamics and function is emerging as a point of convergence in neurodegeneration. Interestingly, α-synuclein and microtubules have been shown to interact and mediate cross-talks with other intracellular structures. This is supported by an increasing amount of evidence ranging from their direct interaction to the engagement of in-common partners and culminating with their respective impact on microtubule-dependent neuronal functions. Last, but not least, it is becoming even more clear that α-synuclein and tubulin work synergically towards pathological aggregation, ultimately resulting in neurodegeneration. In this respect, we supply a novel perspective towards the understanding of α-synuclein biology and, most importantly, of the link between α-synuclein with microtubule cytoskeleton and its impact for neurodegeneration and future development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy; Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | | | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy; Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
20
|
Kim JR. Oligomerization by co-assembly of β-amyloid and α-synuclein. Front Mol Biosci 2023; 10:1153839. [PMID: 37021111 PMCID: PMC10067735 DOI: 10.3389/fmolb.2023.1153839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Aberrant self-assembly of an intrinsically disordered protein is a pathological hallmark of protein misfolding diseases, such as Alzheimer's and Parkinson's diseases (AD and PD, respectively). In AD, the 40-42 amino acid-long extracellular peptide, β-amyloid (Aβ), self-assembles into oligomers, which eventually aggregate into fibrils. A similar self-association of the 140 amino acid-long intracellular protein, α-synuclein (αS), is responsible for the onset of PD pathology. While Aβ and αS are primarily extracellular and intracellular polypeptides, respectively, there is evidence of their colocalization and pathological overlaps of AD and PD. This evidence has raised the likelihood of synergistic, toxic protein-protein interactions between Aβ and αS. This mini review summarizes the findings of studies on Aβ-αS interactions related to enhanced oligomerization via co-assembly, aiming to provide a better understanding of the complex biology behind AD and PD and common pathological mechanisms among the major neurodegenerative diseases.
Collapse
|
21
|
Leitão AD, Spencer B, Sarsoza F, Ngolab J, Amalraj J, Masliah E, Wu C, Rissman RA. Hippocampal Reduction of α-Synuclein via RNA Interference Improves Neuropathology in Alzheimer's Disease Mice. J Alzheimers Dis 2023; 95:349-361. [PMID: 37522208 PMCID: PMC10578232 DOI: 10.3233/jad-230232] [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] [Accepted: 06/23/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) cases are often characterized by the pathological accumulation of α-synuclein (α-syn) in addition to amyloid-β (Aβ) and tau hallmarks. The role of α-syn has been extensively studied in synucleinopathy disorders, but less so in AD. Recent studies have shown that α-syn may also play a role in AD and its downregulation may be protective against the toxic effects of Aβ accumulation. OBJECTIVE We hypothesized that selectively knocking down α-syn via RNA interference improves the neuropathological and biochemical findings in AD mice. METHODS Here we used amyloid precursor protein transgenic (APP-Tg) mice to model AD and explore pathologic and behavioral phenotypes with knockdown of α-syn using RNA interference. We selectively reduced α-syn levels by stereotaxic bilateral injection of either LV-shRNA α-syn or LV-shRNA-luc (control) into the hippocampus of AD mice. RESULTS We found that downregulation of α-syn results in significant reduction in the number of Aβ plaques. In addition, mice treated with LV-shRNA α-syn had amelioration of abnormal microglial activation (Iba1) and astrocytosis (GFAP) phenotypes in AD mice. CONCLUSION Our data suggests a novel link between Aβ and α-syn pathology as well as a new therapeutic angle for targeting AD.
Collapse
Affiliation(s)
- André D.G. Leitão
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Brian Spencer
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Floyd Sarsoza
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| | - Jennifer Ngolab
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jessica Amalraj
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Robert A. Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Physiology and Neuroscience, Alzheimer’s Therapeutic Research Institute of the Keck School of Medicine of the University of Southern California, San Diego, CA, USA
- VA San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
22
|
Jayaraj R, Azimullah S, Parekh KA, Ojha SK, Beiram R. Effect of citronellol on oxidative stress, neuroinflammation and autophagy pathways in an in vivo model of Parkinson's disease. Heliyon 2022; 8:e11434. [DOI: 10.1016/j.heliyon.2022.e11434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/23/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
|
23
|
Dhakal S, Robang AS, Bhatt N, Puangmalai N, Fung L, Kayed R, Paravastu AK, Rangachari V. Distinct neurotoxic TDP-43 fibril polymorphs are generated by heterotypic interactions with α-Synuclein. J Biol Chem 2022; 298:102498. [PMID: 36116552 PMCID: PMC9587012 DOI: 10.1016/j.jbc.2022.102498] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Amyloid aggregates of specific proteins constitute important pathological hallmarks in many neurodegenerative diseases, defining neuronal degeneration and disease onset. Recently, increasing numbers of patients show comorbidities and overlaps between multiple neurodegenerative diseases, presenting distinct phenotypes. Such overlaps are often accompanied by colocalizations of more than one amyloid protein, prompting the question of whether direct interactions between different amyloid proteins could generate heterotypic amyloids. To answer this question, we investigated the effect of α-synuclein (αS) on the DNA-binding protein TDP-43 aggregation inspired by their coexistence in pathologies such as Lewy body dementia and limbic predominant age-related TDP-43 encephalopathy. We previously showed αS and prion-like C-terminal domain (PrLD) of TDP-43 synergistically interact to generate toxic heterotypic aggregates. Here, we extend these studies to investigate whether αS induces structurally and functionally distinct polymorphs of PrLD aggregates. Using αS-PrLD heterotypic aggregates generated in two different stoichiometric proportions, we show αS can affect PrLD fibril forms. PrLD fibrils show distinctive residue level signatures determined by solid state NMR, dye-binding capability, proteinase K (PK) stability, and thermal stability toward SDS denaturation. Furthremore, by gold nanoparticle labeling and transmission electron microscopy, we show the presence of both αS and PrLD proteins within the same fibrils, confirming the existence of heterotypic amyloid fibrils. We also observe αS and PrLD colocalize in the cytosol of neuroblastoma cells and show that the heterotypic PrLD fibrils selectively induce synaptic dysfunction in primary neurons. These findings establish the existence of heterotypic amyloid and provide a molecular basis for the observed overlap between synucleinopathies and TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Shailendra Dhakal
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA; Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, Texas, USA
| | - Leiana Fung
- Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, Texas, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disorders, University of Texas Medical Branch, Galveston, Texas, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA; Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA.
| |
Collapse
|
24
|
Metabolism and memory: α-synuclein level in children with obesity and children with type 1 diabetes; relation to glucotoxicity, lipotoxicity and executive functions. Int J Obes (Lond) 2022; 46:2040-2049. [PMID: 36153375 PMCID: PMC9584809 DOI: 10.1038/s41366-022-01222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/19/2022] [Accepted: 09/05/2022] [Indexed: 11/08/2022]
Abstract
Abstract
Background/Objectives
Children with obesity and those with type 1diabetes (T1D) exhibit subtle neurocognitive deficits, the mechanism of which remains unknown. α-synuclein plays a fundamental role in neurodegeneration. Moreover, its role in glucose and lipids metabolism is emerging. This study aims to assess whether α-synuclein is correlated with the degree of neurodegeneration in children with obesity and those with T1D in comparison to healthy controls and correlate it to various neurocognitive and metabolic parameters.
Subjects/Methods
Forty children with obesity, 40 children with T1D and 40 matched-healthy controls were assessed for anthropometric measurements and blood-pressure. Cognitive evaluation was performed using Stanford–Binet scale and Barkley Deficits in Executive Functioning (EF) Scale-Children and Adolescents. α-synuclein, fasting lipids and glucose were measured with calculation of the homeostatic model of insulin-resistance and estimated-glucose disposal rate.
Results
Children with obesity and those with T1D had significantly higher α-synuclein (p < 0.001) and total EF percentile (p = 0.001) than controls. α-synuclein was negatively correlated to total IQ (p < 0.001 and p = 0.001), and positively correlated with total EF percentile (p = 0.009 and p = 0.001) and EF symptom count percentile (p = 0.005 and p < 0.001) in children with T1D and obesity, respectively. Multivariate-regression revealed that α-synuclein was independently related to age (p = 0.028), diabetes-duration (p = 0.006), HbA1C% (p = 0.034), total IQ (p = 0.013) and EF symptom count percentile (p = 0.003) among children with T1D, and to diastolic blood-pressure percentile (p = 0.013), waist/hip ratio SDS (p = 0.007), total EF percentile (P = 0.033) and EF symptom count percentile (p < 0.001) in children with obesity.
Conclusion
α-synuclein could have a mechanistic role in neurocognitive deficit among children with obesity and T1D.
Collapse
|
25
|
Synucleinopathy in Amyotrophic Lateral Sclerosis: A Potential Avenue for Antisense Therapeutics? Int J Mol Sci 2022; 23:ijms23169364. [PMID: 36012622 PMCID: PMC9409035 DOI: 10.3390/ijms23169364] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease classified as both a neurodegenerative and neuromuscular disorder. With a complex aetiology and no current cure for ALS, broadening the understanding of disease pathology and therapeutic avenues is required to progress with patient care. Alpha-synuclein (αSyn) is a hallmark for disease in neurodegenerative disorders, such as Parkinson's disease, Lewy body dementia, and multiple system atrophy. A growing body of evidence now suggests that αSyn may also play a pathological role in ALS, with αSyn-positive Lewy bodies co-aggregating alongside known ALS pathogenic proteins, such as SOD1 and TDP-43. This review endeavours to capture the scope of literature regarding the aetiology and development of ALS and its commonalities with "synucleinopathy disorders". We will discuss the involvement of αSyn in ALS and motor neuron disease pathology, and the current theories and strategies for therapeutics in ALS treatment, as well as those targeting αSyn for synucleinopathies, with a core focus on small molecule RNA technologies.
Collapse
|
26
|
Evaluation of Alpha-Synuclein Cerebrospinal Fluid Levels in Several Neurological Disorders. J Clin Med 2022; 11:jcm11113139. [PMID: 35683523 PMCID: PMC9181117 DOI: 10.3390/jcm11113139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Alpha-synuclein (α-syn) is a presynaptic neuronal protein that regulates several neuronal functions. In recent decades, the role of α-syn as a biomarker of neurodegenerative diseases has been explored, especially in synucleinopathies. However, only a few studies have assessed its role as biomarker in other neurological disorders. The aim of the study was to evaluate cerebrospinal fluid (CSF) α-syn levels in several neurological disorders; (2) Methods: We measured CSF α-syn levels by a commercial ELISA kit in 158 patients classified in the following group: controls, Alzheimer’s Disease (AD), cerebrovascular diseases, inflammatory central nervous system diseases, other neurological diseases, Parkinson’s Disease (PD), and peripheral neuropathy; (3) Results: Patients with PD showed the lowest and patients with AD the highest levels of CSF α-syn (1372 vs. 2912 pg/mL, respectively, p < 0.001). In AD patients, α-syn levels were significantly associated with tau proteins; (4) Conclusions: α-syn could represent a biomarker of neurodegenerative diseases.
Collapse
|
27
|
Just MK, Gram H, Theologidis V, Jensen PH, Nilsson KPR, Lindgren M, Knudsen K, Borghammer P, Van Den Berge N. Alpha-Synuclein Strain Variability in Body-First and Brain-First Synucleinopathies. Front Aging Neurosci 2022; 14:907293. [PMID: 35693346 PMCID: PMC9178288 DOI: 10.3389/fnagi.2022.907293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Pathogenic alpha-synuclein (asyn) aggregates are a defining feature of neurodegenerative synucleinopathies, which include Parkinson's disease, Lewy body dementia, pure autonomic failure and multiple system atrophy. Early accurate differentiation between these synucleinopathies is challenging due to the highly heterogeneous clinical profile at early prodromal disease stages. Therefore, diagnosis is often made in late disease stages when a patient presents with a broad range of motor and non-motor symptoms easing the differentiation. Increasing data suggest the clinical heterogeneity seen in patients is explained by the presence of distinct asyn strains, which exhibit variable morphologies and pathological functions. Recently, asyn seed amplification assays (PMCA and RT-QuIC) and conformation-specific ligand assays have made promising progress in differentiating between synucleinopathies in prodromal and advanced disease stages. Importantly, the cellular environment is known to impact strain morphology. And, asyn aggregate pathology can propagate trans-synaptically along the brain-body axis, affecting multiple organs and propagating through multiple cell types. Here, we present our hypothesis that the changing cellular environments, an asyn seed may encounter during its brain-to-body or body-to-brain propagation, may influence the structure and thereby the function of the aggregate strains developing within the different cells. Additionally, we aim to review strain characteristics of the different synucleinopathies in clinical and preclinical studies. Future preclinical animal models of synucleinopathies should investigate if asyn strain morphology is altered during brain-to-body and body-to-brain spreading using these seeding amplification and conformation-specific assays. Such findings would greatly deepen our understanding of synucleinopathies and the potential link between strain and phenotypic variability, which may enable specific diagnosis of different synucleinopathies in the prodromal phase, creating a large therapeutic window with potential future applications in clinical trials and personalized therapeutics.
Collapse
Affiliation(s)
- Mie Kristine Just
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Hjalte Gram
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Vasileios Theologidis
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Poul Henning Jensen
- Department of Biomedicine, DANDRITE-Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - K. Peter R. Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Mikael Lindgren
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karoline Knudsen
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Nathalie Van Den Berge
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
28
|
Gramotnev DK, Gramotnev G, Gramotnev A, Summers MJ. Path analysis of biomarkers for cognitive decline in early Parkinson’s disease. PLoS One 2022; 17:e0268379. [PMID: 35560326 PMCID: PMC9106174 DOI: 10.1371/journal.pone.0268379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Clinical and biochemical diversity of Parkinson’s disease (PD) and numerous demographic, clinical, and pathological measures influencing cognitive function and its decline in PD create problems with the determination of effects of individual measures on cognition in PD. This is particularly the case where these measures significantly interrelate with each other producing intricate networks of direct and indirect effects on cognition. Here, we use generalized structural equation modelling (GSEM) to identify and characterize significant paths for direct and indirect effects of 14 baseline measures on global cognition in PD at baseline and at 4 years later. We consider 269 drug-naïve participants from the Parkinson’s Progression Marker Initiative database, diagnosed with idiopathic PD and observed for at least 4 years after baseline. Two GSEM networks are derived, highlighting the possibility of at least two different molecular pathways or two different PD sub-types, with either CSF p-tau181 or amyloid beta (1–42) being the primary protein variables potentially driving progression of cognitive decline. The models provide insights into the interrelations between the 14 baseline variables, and determined their total effects on cognition in early PD. High CSF amyloid concentrations (> 500 pg/ml) are associated with nearly full protection against cognitive decline in early PD in the whole range of baseline age between 40 and 80 years, and irrespectively of whether p-tau181 or amyloid beta (1–42) are considered as the primary protein variables. The total effect of depression on cognition is shown to be strongly amplified by PD, but not at the time of diagnosis or at prodromal stages. CSF p-tau181 protein could not be a reliable indicator of cognitive decline because of its significantly heterogeneous effects on cognition. The outcomes will enable better understanding of the roles of the clinical and pathological measures and their mutual effects on cognition in early PD.
Collapse
Affiliation(s)
| | - Galina Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
| | - Alexandra Gramotnev
- Research and Data Analysis Centre, Brisbane, Queensland, Australia
- Sunshine Coast Mind & Neuroscience – Thompson Institute, University of the Sunshine Coast, Birtinya, Queensland, Australia
| | - Mathew J. Summers
- School of Health and Behavioural Science, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
29
|
Synhaivska O, Bhattacharya S, Campioni S, Thompson D, Nirmalraj PN. Single-Particle Resolution of Copper-Associated Annular α-Synuclein Oligomers Reveals Potential Therapeutic Targets of Neurodegeneration. ACS Chem Neurosci 2022; 13:1410-1421. [PMID: 35414168 PMCID: PMC9073932 DOI: 10.1021/acschemneuro.2c00021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
![]()
Metal ions stabilize
protein–protein interactions and can
modulate protein aggregation. Here, using liquid-based atomic force
microscopy and molecular dynamics simulations, we study the concentration-dependent
effect of Cu2+ ions on the aggregation pathway of α-synuclein
(α-Syn) proteins, which play a key role in the pathology of
Parkinson’s disease. The full spectrum of α-Syn aggregates
in the presence and absence of Cu2+ ions from monomers
to mature fibrils was resolved and quantified at the gold–water
interface. Raman spectroscopy confirmed the atomic force microscopy
(AFM) findings on the heterogeneity in aggregated states of α-Syn.
The formation of annular oligomers was exclusively detected upon incubating
α-Syn with Cu2+ ions. Our findings emphasize the
importance of targeting annular α-Syn protein oligomers for
therapeutic intervention and their potential role as biomarkers for
early detection and monitoring progression of neurodegeneration.
Collapse
Affiliation(s)
- Olena Synhaivska
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| | - Shayon Bhattacharya
- Department of Physics, Bernal Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Silvia Campioni
- Functional Materials Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| | - Damien Thompson
- Department of Physics, Bernal Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| |
Collapse
|
30
|
Beatino MF, De Luca C, Campese N, Belli E, Piccarducci R, Giampietri L, Martini C, Perugi G, Siciliano G, Ceravolo R, Vergallo A, Hampel H, Baldacci F. α-synuclein as an emerging pathophysiological biomarker of Alzheimer's disease. Expert Rev Mol Diagn 2022; 22:411-425. [PMID: 35443850 DOI: 10.1080/14737159.2022.2068952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION α-syn aggregates represent the pathological hallmark of synucleinopathies as well as a frequent copathology (almost 1/3 of cases) in AD. Recent research indicates a potential role of α-syn species, measured in CSF with conventional analytical techniques, in the differential diagnosis between AD and synucleinopathies (such as DLB). Pioneering studies report the detection of α-syn in blood, however, conclusive investigations are controversial. Ultrasensitive seed amplification techniques, enabling the selective quantification of α-syn seeds, may represent an effective solution to identify the α-syn component in AD and facilitate a biomarker-guided stratification. AREAS COVERED We performed a PubMed-based review of the latest findings on α-syn-related biomarkers for AD, focusing on bodily fluids. A dissertation on the role of ultrasensitive seed amplification assays, detecting α-syn seeds from different biological samples, was conducted. EXPERT OPINION α-syn may contribute to progressive AD neurodegeneration through cross-seeding especially with tau protein. Ultrasensitive seed amplification techniques may support a biomarker-drug co-development pathway and may be a pathophysiological candidate biomarker for the evolving ATX(N) system to classify AD and the spectrum of primary NDDs. This would contribute to a precise approach to AD, aimed at implementing disease-modifying treatments.
Collapse
Affiliation(s)
| | - Ciro De Luca
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicole Campese
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisabetta Belli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Giulio Perugi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Vergallo
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| | - Harald Hampel
- GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,GRC N° 21, Alzheimer Precision Medicine (APM), AP-HP, Sorbonne University, Pitié-Salpêtrière Hospital, Boulevard De l'Hôpital, Paris, France
| |
Collapse
|
31
|
Insights into the inhibitory mechanism of skullcapflavone II against α-synuclein aggregation and its mediated cytotoxicity. Int J Biol Macromol 2022; 209:426-440. [PMID: 35398391 DOI: 10.1016/j.ijbiomac.2022.03.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 11/21/2022]
Abstract
The dangerous self-assembled and infectious seeds of α-synuclein (αSN) play primary roles in Parkinson's disease. Accordingly, the inhibition of αSN fibrillation and elimination of toxic aggregates are the main therapeutic strategies. Skullcapflavone II (S.FII), a compound isolated from S. pinnatifida, has shown multiple neuroprotective features. Herein, we demonstrated that S.FII inhibited αSN aggregation with IC50 of 7.2 μM. It increased nucleation time and decreased fibril elongation rate and the species formed in the presence of S.FII were unable to act as seeds. Additionally, S.FII inhibited both secondary nucleation and seeding of αSN and disaggregated the mature preformed fibrils as well. The species formed in the presence of S.FII showed less toxicity. It also preserved neurite length and dopamine content of SH-SY5Y cells and attenuated the inflammatory responses in mixed glial cells. The Localized Surface Plasmon Resonance (LSPR) analysis indicated that S.FII interacts with αSN. Docking simulation studies on αSN fibrils revealed that S.FII could interact with the key residues of the salt bridges and glutamine ladder, which might lead to the destruction of fibril's structures. We also showed that S.FII passes through the blood-brain barrier in vitro and in vivo. Overall, these findings elucidate the neuroprotective roles of S.FII in reducing αSN pathogenicity.
Collapse
|
32
|
Sengupta U, Kayed R. Amyloid β, Tau, and α-Synuclein aggregates in the pathogenesis, prognosis, and therapeutics for neurodegenerative diseases. Prog Neurobiol 2022; 214:102270. [DOI: 10.1016/j.pneurobio.2022.102270] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
|
33
|
Passive Immunization in Alpha-Synuclein Preclinical Animal Models. Biomolecules 2022; 12:biom12020168. [PMID: 35204668 PMCID: PMC8961624 DOI: 10.3390/biom12020168] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 12/20/2022] Open
Abstract
Alpha-synucleinopathies include Parkinson’s disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy. These are all progressive neurodegenerative diseases that are characterized by pathological misfolding and accumulation of the protein alpha-synuclein (αsyn) in neurons, axons or glial cells in the brain, but also in other organs. The abnormal accumulation and propagation of pathogenic αsyn across the autonomic connectome is associated with progressive loss of neurons in the brain and peripheral organs, resulting in motor and non-motor symptoms. To date, no cure is available for synucleinopathies, and therapy is limited to symptomatic treatment of motor and non-motor symptoms upon diagnosis. Recent advances using passive immunization that target different αsyn structures show great potential to block disease progression in rodent studies of synucleinopathies. However, passive immunotherapy in clinical trials has been proven safe but less effective than in preclinical conditions. Here we review current achievements of passive immunotherapy in animal models of synucleinopathies. Furthermore, we propose new research strategies to increase translational outcome in patient studies, (1) by using antibodies against immature conformations of pathogenic αsyn (monomers, post-translationally modified monomers, oligomers and protofibrils) and (2) by focusing treatment on body-first synucleinopathies where damage in the brain is still limited and effective immunization could potentially stop disease progression by blocking the spread of pathogenic αsyn from peripheral organs to the brain.
Collapse
|
34
|
Zhao X, Hasan S, Liou B, Lin Y, Sun Y, Liu C. Analysis of the Biomarkers for Neurodegenerative Diseases in Aged Progranulin Deficient Mice. Int J Mol Sci 2022; 23:629. [PMID: 35054815 PMCID: PMC8775568 DOI: 10.3390/ijms23020629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022] Open
Abstract
Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and β-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.
Collapse
Affiliation(s)
- Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Sadaf Hasan
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Yi Lin
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Ying Sun
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
35
|
Goulding SR, Anantha J, Collins LM, Sullivan AM, O'Keeffe GW. Growth differentiation factor 5: a neurotrophic factor with neuroprotective potential in Parkinson's disease. Neural Regen Res 2022; 17:38-44. [PMID: 34100424 PMCID: PMC8451580 DOI: 10.4103/1673-5374.314290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/08/2021] [Accepted: 01/24/2021] [Indexed: 11/04/2022] Open
Abstract
Parkinson's disease is the most common movement disorder worldwide, affecting over 6 million people. It is an age-related disease, occurring in 1% of people over the age of 60, and 3% of the population over 80 years. The disease is characterized by the progressive loss of midbrain dopaminergic neurons from the substantia nigra, and their axons, which innervate the striatum, resulting in the characteristic motor and non-motor symptoms of Parkinson's disease. This is paralleled by the intracellular accumulation of α-synuclein in several regions of the nervous system. Current therapies are solely symptomatic and do not stop or slow disease progression. One promising disease-modifying strategy to arrest the loss of dopaminergic neurons is the targeted delivery of neurotrophic factors to the substantia nigra or striatum, to protect the remaining dopaminergic neurons of the nigrostriatal pathway. However, clinical trials of two well-established neurotrophic factors, glial cell line-derived neurotrophic factor and neurturin, have failed to meet their primary end-points. This failure is thought to be at least partly due to the downregulation by α-synuclein of Ret, the common co-receptor of glial cell line-derived neurorophic factor and neurturin. Growth/differentiation factor 5 is a member of the bone morphogenetic protein family of neurotrophic factors, that signals through the Ret-independent canonical Smad signaling pathway. Here, we review the evidence for the neurotrophic potential of growth/differentiation factor 5 in in vitro and in vivo models of Parkinson's disease. We discuss new work on growth/differentiation factor 5's mechanisms of action, as well as data showing that viral delivery of growth/differentiation factor 5 to the substantia nigra is neuroprotective in the α-synuclein rat model of Parkinson's disease. These data highlight the potential for growth/differentiation factor 5 as a disease-modifying therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Susan R. Goulding
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Jayanth Anantha
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Louise M. Collins
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Das TK, Blasco-Conesa MP, Korf J, Honarpisheh P, Chapman MR, Ganesh BP. Bacterial Amyloid Curli Associated Gut Epithelial Neuroendocrine Activation Predominantly Observed in Alzheimer's Disease Mice with Central Amyloid-β Pathology. J Alzheimers Dis 2022; 88:191-205. [PMID: 35527554 PMCID: PMC9583710 DOI: 10.3233/jad-220106] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Substantial evidence from recent research suggests an influential and underappreciated force in Alzheimer's disease (AD) pathogenesis: the pathological signals originate from outside the brain. Pathogenic bacteria produce amyloid-like proteins "curli" that form biofilms and show functional similarities to human amyloid-β (Aβ). These proteins may contribute to neurological disease progression via signaling cascade from the gut to the brain. OBJECTIVE We propose that curli causes neuroendocrine activation from the gut to brain that promotes central Aβ pathology. METHODS PGP9.5 and TLR2 levels in response to curli in the lumen of Tg2576 AD mice were analyzed by immunohistochemical and qRT-PCR analysis. Western blot and human 3D in vitro enteroids culture systems were also used. 16S rRNA gene sequencing was used to investigate bacterial dysbiosis. RESULTS We found significant increase in bacterial-amyloid curli with elevated TLR2 at the mRNA level in the pre- and symptomatic Tg-AD gut compared to littermate WT controls. This data associates with increased gram-positive bacterial colonization in the ileum of the symptomatic AD mice. We found fundamental evidence for vagus nerve activation in response to bacterial curli. Neuroendocrine marker PGP9.5 was significantly elevated in the gut epithelium of symptomatic AD mice, and this was colocalized with increased TLR2 expression. Enteroids, 3D-human ileal mini-gut monolayer in vitro model system also revealed increase levels of TLR2 upon stimulation with purified bacterial curli fibrils. CONCLUSION These findings reveal the importance of pathological changes within the gut-vagus-brain signaling in response to luminal bacterial amyloid that might play a vital role in central Aβ pathogenesis seen in the AD brain.
Collapse
Affiliation(s)
- Tushar K. Das
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maria P. Blasco-Conesa
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Janelle Korf
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pedram Honarpisheh
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew R. Chapman
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Bhanu P. Ganesh
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA,Correspondence to: Bhanu Priya Ganesh, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA. Tel.: +1 713 500 7429;
| |
Collapse
|
37
|
Kim H, Jung HR, Kim JB, Kim DJ. Autonomic Dysfunction in Sleep Disorders: From Neurobiological Basis to Potential Therapeutic Approaches. J Clin Neurol 2022; 18:140-151. [PMID: 35274834 PMCID: PMC8926769 DOI: 10.3988/jcn.2022.18.2.140] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022] Open
Abstract
Sleep disorder has been portrayed as merely a common dissatisfaction with sleep quality and quantity. However, sleep disorder is actually a medical condition characterized by inconsistent sleep patterns that interfere with emotional dynamics, cognitive functioning, and even physical performance. This is consistent with sleep abnormalities being more common in patients with autonomic dysfunction than in the general population. The autonomic nervous system coordinates various visceral functions ranging from respiration to neuroendocrine secretion in order to maintain homeostasis of the body. Because the cell population and efferent signals involved in autonomic regulation are spatially adjacent to those that regulate the sleep-wake system, sleep architecture and autonomic coordination exert effects on each other, suggesting the presence of a bidirectional relationship in addition to shared pathology. The primary goal of this review is to highlight the bidirectional and shared relationship between sleep and autonomic regulation. It also introduces the effects of autonomic dysfunction on insomnia, breathing disorders, central disorders of hypersomnolence, parasomnias, and movement disorders. This information will assist clinicians in determining how neuromodulation can have the greatest therapeutic effects in patients with sleep disorders.
Collapse
Affiliation(s)
- Hakseung Kim
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea
| | - Hee Ra Jung
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Jung Bin Kim
- Department of Neurology, Korea University College of Medicine, Seoul, Korea
| | - Dong-Joo Kim
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea
- Department of Neurology, Korea University College of Medicine, Seoul, Korea
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- NeuroTx, Co., Ltd., Seoul, Korea
| |
Collapse
|
38
|
Neuroimmune contributions to Alzheimer's disease: a focus on human data. Mol Psychiatry 2022; 27:3164-3181. [PMID: 35668160 PMCID: PMC9168642 DOI: 10.1038/s41380-022-01637-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
The past decade has seen the convergence of a series of new insights that arose from genetic and systems analyses of Alzheimer's disease (AD) with a wealth of epidemiological data from a variety of fields; this resulted in renewed interest in immune responses as important, potentially causal components of AD. Here, we focus primarily on a review of human data which has recently yielded a set of robust, reproducible results that exist in a much larger universe of conflicting reports stemming from small studies with important limitations in their study design. Thus, we are at an important crossroads in efforts to first understand at which step of the long, multiphasic course of AD a given immune response may play a causal role and then modulate this response to slow or block the pathophysiology of AD. We have a wealth of new experimental tools, analysis methods, and capacity to sample human participants at large scale longitudinally; these resources, when coupled to a foundation of reproducible results and novel study designs, will enable us to monitor human immune function in the CNS at the level of complexity that is required while simultaneously capturing the state of the peripheral immune system. This integration of peripheral and central perturbations in immune responses results in pathologic responses in the central nervous system parenchyma where specialized cellular microenvironments composed of multiple cell subtypes respond to these immune perturbations as well as to environmental exposures, comorbidities and the impact of the advancing life course. Here, we offer an overview that seeks to illustrate the large number of interconnecting factors that ultimately yield the neuroimmune component of AD.
Collapse
|
39
|
Parkin beyond Parkinson’s Disease—A Functional Meaning of Parkin Downregulation in TDP-43 Proteinopathies. Cells 2021; 10:cells10123389. [PMID: 34943897 PMCID: PMC8699658 DOI: 10.3390/cells10123389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Parkin and PINK1 are key regulators of mitophagy, an autophagic pathway for selective elimination of dysfunctional mitochondria. To this date, parkin depletion has been associated with recessive early onset Parkinson’s disease (PD) caused by loss-of-function mutations in the PARK2 gene, while, in sporadic PD, the activity and abundance of this protein can be compromised by stress-related modifications. Intriguingly, research in recent years has shown that parkin depletion is not limited to PD but is also observed in other neurodegenerative diseases—especially those characterized by TDP-43 proteinopathies, such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Here, we discuss the evidence of parkin downregulation in these disease phenotypes, its emerging connections with TDP-43, and its possible functional implications.
Collapse
|
40
|
Visanji NP, Kovacs GG, Lang AE. The Discovery of α-Synuclein in Lewy Pathology of Parkinson's Disease: The Inspiration of a Revolution. Mov Disord Clin Pract 2021; 8:1189-1193. [PMID: 34765684 DOI: 10.1002/mdc3.13312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 11/09/2022] Open
Affiliation(s)
- Naomi P Visanji
- Edmond J. Safra program in Parkinson's disease and the Morton and Gloria Shulman Movement Disorders Clinic Toronto Western Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
| | - Gabor G Kovacs
- Edmond J. Safra program in Parkinson's disease and the Morton and Gloria Shulman Movement Disorders Clinic Toronto Western Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada.,Tanz Centre for Research in Neurodegenerative Disease University of Toronto Toronto Ontario Canada
| | - Anthony E Lang
- Edmond J. Safra program in Parkinson's disease and the Morton and Gloria Shulman Movement Disorders Clinic Toronto Western Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
| |
Collapse
|
41
|
Dubackic M, Idini I, Lattanzi V, Liu Y, Martel A, Terry A, Haertlein M, Devos JM, Jackson A, Sparr E, Linse S, Olsson U. On the Cluster Formation of α-Synuclein Fibrils. Front Mol Biosci 2021; 8:768004. [PMID: 34738016 PMCID: PMC8560691 DOI: 10.3389/fmolb.2021.768004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/30/2021] [Indexed: 12/05/2022] Open
Abstract
The dense accumulation of α-Synuclein fibrils in neurons is considered to be strongly associated with Parkinson’s disease. These intracellular inclusions, called Lewy bodies, also contain significant amounts of lipids. To better understand such accumulations, it should be important to study α-Synuclein fibril formation under conditions where the fibrils lump together, mimicking what is observed in Lewy bodies. In the present study, we have therefore investigated the overall structural arrangements of α-synuclein fibrils, formed under mildly acidic conditions, pH = 5.5, in pure buffer or in the presence of various model membrane systems, by means of small-angle neutron scattering (SANS). At this pH, α-synuclein fibrils are colloidally unstable and aggregate further into dense clusters. SANS intensities show a power law dependence on the scattering vector, q, indicating that the clusters can be described as mass fractal aggregates. The experimentally observed fractal dimension was d = 2.6 ± 0.3. We further show that this fractal dimension can be reproduced using a simple model of rigid-rod clusters. The effect of dominatingly attractive fibril-fibril interactions is discussed within the context of fibril clustering in Lewy body formation.
Collapse
Affiliation(s)
- Marija Dubackic
- Division of Physical Chemistry, Department of Chemistry, Lund University, Lund, Sweden
| | - Ilaria Idini
- Division of Physical Chemistry, Department of Chemistry, Lund University, Lund, Sweden
| | - Veronica Lattanzi
- Division of Physical Chemistry, Department of Chemistry, Lund University, Lund, Sweden.,Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Yun Liu
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD, United States.,Chemical and Biomolecular Engineering Department, University of Delaware, Newark, DE, United States
| | | | - Ann Terry
- ISIS Neutron and Muon Source, Harwell Oxford, Didcot, United Kingdom.,Max IV Laboratory, Lund University, Lund, Sweden
| | | | | | - Andrew Jackson
- Division of Physical Chemistry, Department of Chemistry, Lund University, Lund, Sweden.,European Spallation Source, Lund, Sweden
| | - Emma Sparr
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Sara Linse
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Ulf Olsson
- Division of Physical Chemistry, Department of Chemistry, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Jellinger KA, Wenning GK, Stefanova N. Is Multiple System Atrophy a Prion-like Disorder? Int J Mol Sci 2021; 22:10093. [PMID: 34576255 PMCID: PMC8472631 DOI: 10.3390/ijms221810093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive, fatal neurodegenerative disease of uncertain aetiology that belongs to the family of α-synucleinopathies. It clinically presents with parkinsonism, cerebellar, autonomic, and motor impairment in variable combinations. Pathological hallmarks are fibrillary α-synuclein (αSyn)-rich glial cytoplasmic inclusions (GCIs) mainly involving oligodendroglia and to a lesser extent neurons, inducing a multisystem neurodegeneration, glial activation, and widespread demyelinization. The neuronal αSyn pathology of MSA has molecular properties different from Lewy bodies in Parkinson's disease (PD), both of which could serve as a pool of αSyn (prion) seeds that could initiate and drive the pathogenesis of synucleinopathies. The molecular cascade leading to the "prion-like" transfer of "strains" of aggregated αSyn contributing to the progression of the disease is poorly understood, while some presented evidence that MSA is a prion disease. However, this hypothesis is difficult to reconcile with postmortem analysis of human brains and the fact that MSA-like pathology was induced by intracerebral inoculation of human MSA brain homogenates only in homozygous mutant 53T mice, without production of disease-specific GCIs, or with replication of MSA prions in primary astrocyte cultures from transgenic mice expressing human αSyn. Whereas recent intrastriatal injection of Lewy body-derived or synthetic human αSyn fibrils induced PD-like pathology including neuronal αSyn aggregates in macaques, no such transmission of αSyn pathology in non-human primates by MSA brain lysate has been reported until now. Given the similarities between αSyn and prions, there is a considerable debate whether they should be referred to as "prions", "prion-like", "prionoids", or something else. Here, the findings supporting the proposed nature of αSyn as a prion and its self-propagation through seeding as well as the transmissibility of neurodegenerative disorders are discussed. The proof of disease causation rests on the concordance of scientific evidence, none of which has provided convincing evidence for the classification of MSA as a prion disease or its human transmission until now.
Collapse
Affiliation(s)
| | - Gregor K. Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| |
Collapse
|
43
|
Quint WH, Matečko-Burmann I, Schilcher I, Löffler T, Schöll M, Burmann BM, Vogels T. Bispecific Tau Antibodies with Additional Binding to C1q or Alpha-Synuclein. J Alzheimers Dis 2021; 80:813-829. [PMID: 33579845 DOI: 10.3233/jad-201334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) and other tauopathies are neurodegenerative disorders characterized by cellular accumulation of aggregated tau protein. Tau pathology within these disorders is accompanied by chronic neuroinflammation, such as activation of the classical complement pathway by complement initiation factor C1q. Additionally, about half of the AD cases present with inclusions composed of aggregated alpha-synuclein called Lewy bodies. Lewy bodies in disorders such as Parkinson's disease and Lewy body dementia also frequently occur together with tau pathology. OBJECTIVE Immunotherapy is currently the most promising treatment strategy for tauopathies. However, the presence of multiple pathological processes within tauopathies makes it desirable to simultaneously target more than one disease pathway. METHODS Herein, we have developed three bispecific antibodies based on published antibody binding region sequences. One bispecific antibody binds to tau plus alpha-synuclein and two bispecific antibodies bind to tau plus C1q. RESULTS Affinity of the bispecific antibodies to their targets compared to their monospecific counterparts ranged from nearly identical to one order of magnitude lower. All bispecific antibodies retained binding to aggregated protein in patient-derived brain sections. The bispecific antibodies also retained their ability to inhibit aggregation of recombinant tau, regardless of whether the tau binding sites were in IgG or scFv format. Mono- and bispecific antibodies inhibited cellular seeding induced by AD-derived pathological tau with similar efficacy. Finally, both Tau-C1q bispecific antibodies completely inhibited the classical complement pathway. CONCLUSION Bispecific antibodies that bind to multiple pathological targets may therefore present a promising approach to treat tauopathies and other neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Irena Matečko-Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Tina Löffler
- QPS Austria GmbH, Neuropharmacology, Grambach, Austria
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| | - Björn Marcus Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Vogels
- Maptimmune BV, The Hague, The Netherlands.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| |
Collapse
|
44
|
Poewe W, Volc D, Seppi K, Medori R, Lührs P, Kutzelnigg A, Djamshidian A, Thun-Hohenstein C, Meissner WG, Rascol O, Schneeberger A, Staffler G, Poewe W, Seppi K, Djamshidian A, deMarzi R, Heim B, Mangesius S, Stolz R, Wachowicz K, Volc D, Thun-Hohenstein C, Riha C, Schneeberger A, Bürger V, Galabova G. Safety and Tolerability of Active Immunotherapy Targeting α-Synuclein with PD03A in Patients with Early Parkinson's Disease: A Randomized, Placebo-Controlled, Phase 1 Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:1079-1089. [PMID: 34092654 PMCID: PMC8461711 DOI: 10.3233/jpd-212594] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: Immunotherapies targeting α-synuclein aim to limit its extracellular spread in the brain and prevent progression of pathology in Parkinson’s disease (PD). PD03A is a specific active immunotherapy (SAIT) involving immunization with a short peptide formulation. Objective: This phase 1 study characterized the safety and tolerability of PD03A in patients with early PD. A key secondary objective was to evaluate immunological activity following immunization. Methods: This was a phase 1 study of two different doses of PD03A versus placebo in PD patients. Patients were randomized (1:1:1) to receive four priming plus one booster vaccination of PD03A 15μg, PD03A 75μg or placebo and were followed for 52 weeks. Results: Overall, 36 patients were randomized, of which 35 received five immunizations and completed the study. All patients experienced at least one adverse event. Transient local injection site reactions affected all but two patients; otherwise most AEs were considered unrelated to study treatment. A substantial IgG antibody response against PD03 was observed with a maximum titer achieved at Week-12. Differences in titers between both active groups versus placebo were statistically significant from the second immunization at Week-8 until Week-52. Conclusion: The safety profile and positive antibody response of PD03A supports the further development of active immunotherapeutic approaches for the treatment of PD.
Collapse
Affiliation(s)
- Werner Poewe
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Dieter Volc
- PROSENEX Study Center at Privatklinik Confraternitaet, Vienna, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | - Atbin Djamshidian
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Wassilios G Meissner
- Service de Neurologie, CRMR Atrophie Multisystématisée, CHU Bordeaux and Université Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Olivier Rascol
- Toulouse Parkinson Expert Center, Departments of Neurosciences and Clinical Pharmacology, Centre d'Investigation Clinique de Toulouse CIC1436, NS-Park/FCRIN Network, and NeuroToul COEN Center, University Hospital of Toulouse, INSERM, University of Toulouse, Toulouse, France
| | | | | | | | | | - Werner Poewe
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Atbin Djamshidian
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Roberto deMarzi
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Beatrice Heim
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Raphaela Stolz
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Dieter Volc
- PROSENEX Study Center at Privatklinik Confraternitaet, Vienna, Austria
| | | | - Constanze Riha
- PROSENEX Study Center at Privatklinik Confraternitaet, Vienna, Austria
| | | | | | | |
Collapse
|
45
|
Dai L, Wang J, He M, Xiong M, Tian Y, Liu C, Zhang Z. Lovastatin Alleviates α-Synuclein Aggregation and Phosphorylation in Cellular Models of Synucleinopathy. Front Mol Neurosci 2021; 14:682320. [PMID: 34381332 PMCID: PMC8350347 DOI: 10.3389/fnmol.2021.682320] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases. Pathologically, it is characterized by the aberrant aggregation of α-synuclein (α-syn) in neurons. Clinical evidence shows that patients with hypercholesterolemia are more likely to get PD, while lovastatin users have a lower risk of suffering from it. In this study, we investigated the effects of lovastatin on the aggregation and phosphorylation of α-syn in vitro. Our results demonstrate that α-syn preformed fibrils induce the phosphorylation and aggregation of α-syn in HEK293 cells stably transfected with α-syn-GFP and SH-SY5Y cells as well, which could be attenuated by in a concentration-dependent manner. Besides, lovastatin inhibited oxidative stress, histone acetylation, and the activation of casein kinase 2 (CK2). Collectively, lovastatin alleviates α-syn aggregation and phosphorylation in cellular models of synucleinopathy, indicating its potential value of being adopted in the management of PD.
Collapse
Affiliation(s)
- Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiannan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingyang He
- Hubei Provincial Institute for Food Supervision and Test, Wuhan, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ye Tian
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chaoyang Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China.,Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Motyl JA, Strosznajder JB, Wencel A, Strosznajder RP. Recent Insights into the Interplay of Alpha-Synuclein and Sphingolipid Signaling in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22126277. [PMID: 34207975 PMCID: PMC8230587 DOI: 10.3390/ijms22126277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 01/22/2023] Open
Abstract
Molecular studies have provided increasing evidence that Parkinson’s disease (PD) is a protein conformational disease, where the spread of alpha-synuclein (ASN) pathology along the neuraxis correlates with clinical disease outcome. Pathogenic forms of ASN evoke oxidative stress (OS), neuroinflammation, and protein alterations in neighboring cells, thereby intensifying ASN toxicity, neurodegeneration, and neuronal death. A number of evidence suggest that homeostasis between bioactive sphingolipids with opposing function—e.g., sphingosine-1-phosphate (S1P) and ceramide—is essential in pro-survival signaling and cell defense against OS. In contrast, imbalance of the “sphingolipid biostat” favoring pro-oxidative/pro-apoptotic ceramide-mediated changes have been indicated in PD and other neurodegenerative disorders. Therefore, we focused on the role of sphingolipid alterations in ASN burden, as well as in a vast range of its neurotoxic effects. Sphingolipid homeostasis is principally directed by sphingosine kinases (SphKs), which synthesize S1P—a potent lipid mediator regulating cell fate and inflammatory response—making SphK/S1P signaling an essential pharmacological target. A growing number of studies have shown that S1P receptor modulators, and agonists are promising protectants in several neurological diseases. This review demonstrates the relationship between ASN toxicity and alteration of SphK-dependent S1P signaling in OS, neuroinflammation, and neuronal death. Moreover, we discuss the S1P receptor-mediated pathways as a novel promising therapeutic approach in PD.
Collapse
Affiliation(s)
- Joanna A. Motyl
- Department of Hybrid Microbiosystems Engineering, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland; (J.A.M.); (A.W.)
| | - Joanna B. Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego St., 02-106 Warsaw, Poland;
| | - Agnieszka Wencel
- Department of Hybrid Microbiosystems Engineering, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Ks. Trojdena 4 St., 02-109 Warsaw, Poland; (J.A.M.); (A.W.)
| | - Robert P. Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego St., 02-106 Warsaw, Poland
- Correspondence:
| |
Collapse
|
47
|
Sato T, Yajima T, Saijyo S, Shimazaki K, Nishitani T, Hoshika T, Nishitani Y, Ichikawa H, Mizoguchi I, Fukunaga T. Distribution of alpha-synuclein in the rat cranial sensory ganglia, and oro-cervical regions. Ann Anat 2021; 238:151776. [PMID: 34082081 DOI: 10.1016/j.aanat.2021.151776] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/24/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alpha-synuclein (Syn), an unfolded soluble cytosolic protein, is known as a disease-associated protein in the brain. However, little is known about distribution of this protein in the peripheral nervous system. In this study, expression of Syn was investigated in the sensory ganglia of the cranial nerves V, IX and X. METHODS To analyze distribution of Syn and its co-expression with calcitonin gene-related peptide (CGRP) or the transient receptor potential cation channel subfamily V member 1 (TRPV1), immunohistochemical techniques were used in the rat cranial sensory ganglia and their peripheral tissues. RESULTS Syn-immunoreactive (-ir) neurons were abundant in the sensory ganglia of the petrosal (56.7%), jugular (28.3%) and nodose ganglia (82.5%). These neurons had small to medium-sized cell bodies (petrosal, mean ± S.D. = 667.4 ± 310.8 μ m2; jugular, 625.1 ± 318.4 μ m2; nodose, 708.3 ± 248.3 μ m2), and were distributed throughout the ganglia. However, the trigeminal ganglion was mostly free of Syn-ir neurons. By double and triple immunofluorescence staining, Syn-ir neurons co-expressed CGRP and TRPV1 in the petrosal and jugular ganglia. Syn-immunoreactivity was expressed by nerve fibers in the epithelium and taste bud of oral and cervical viscerae. These nerve fibers were abundant in the naso-pharynx, epiglottis and laryngeal vestibule. Some taste bud cells were also immunoreactive for Syn. In addition, Syn-ir nerve fibers were detected in the vicinity of macrophages, dendritic cells and Langerhans cells. CONCLUSIONS Syn was abundant in the visceral sensory neurons but not in somatic sensory neurons. This protein may play a role in nociceptive and chemosensory transduction in the glossopharyngeal and vagal sensory ganglia. It is possible that Syn has a function about the immune mechanism of the upper air way.
Collapse
Affiliation(s)
- Tadasu Sato
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan.
| | - Takehiro Yajima
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Shiori Saijyo
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Kenichiro Shimazaki
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Tomiko Nishitani
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima 890-8544, Japan
| | - Tomohiro Hoshika
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Research Field in Dentistry, Medical and Dental Sciences Area, Research and Education Assembly, Kagoshima University, Kagoshima 890-8544, Japan
| | - Hiroyuki Ichikawa
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Itaru Mizoguchi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Tomohiro Fukunaga
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
48
|
Ray B, Mahalakshmi AM, Tuladhar S, Bhat A, Srinivasan A, Pellegrino C, Kannan A, Bolla SR, Chidambaram SB, Sakharkar MK. "Janus-Faced" α-Synuclein: Role in Parkinson's Disease. Front Cell Dev Biol 2021; 9:673395. [PMID: 34124057 PMCID: PMC8194081 DOI: 10.3389/fcell.2021.673395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/15/2021] [Indexed: 01/03/2023] Open
Abstract
Parkinson's disease (PD) is a pathological condition characterized by the aggregation and the resultant presence of intraneuronal inclusions termed Lewy bodies (LBs) and Lewy neurites which are mainly composed of fibrillar α-synuclein (α-syn) protein. Pathogenic aggregation of α-syn is identified as the major cause of LBs deposition. Several mutations in α-syn showing varied aggregation kinetics in comparison to the wild type (WT) α-syn are reported in PD (A30P, E46K, H 50Q, G51D, A53E, and A53T). Also, the cell-to-cell spread of pathological α-syn plays a significant role in PD development. Interestingly, it has also been suggested that the pathology of PD may begin in the gastrointestinal tract and spread via the vagus nerve (VN) to brain proposing the gut-brain axis of α-syn pathology in PD. Despite multiple efforts, the behavior and functions of this protein in normal and pathological states (specifically in PD) is far from understood. Furthermore, the etiological factors responsible for triggering aggregation of this protein remain elusive. This review is an attempt to collate and present latest information on α-syn in relation to its structure, biochemistry and biophysics of aggregation in PD. Current advances in therapeutic efforts toward clearing the pathogenic α-syn via autophagy/lysosomal flux are also reviewed and reported.
Collapse
Affiliation(s)
- Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Sunanda Tuladhar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Asha Srinivasan
- Division of Nanoscience & Technology, Faculty of Life Sciences, JSS Academy of Higher Education & Research, Mysuru, India
| | - Christophe Pellegrino
- Institut National de la Santé et de la Recherche Médicale, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Anbarasu Kannan
- Department of Protein Chemistry and Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Srinivasa Rao Bolla
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan City, Kazakhstan
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- Special Interest Group – Brain, Behaviour, and Cognitive Neurosciences Research, JSS Academy of Higher Education & Research, Mysuru, India
| | | |
Collapse
|
49
|
Ernest James Phillips T, Maguire E. Phosphoinositides: Roles in the Development of Microglial-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2021; 15:652593. [PMID: 33841102 PMCID: PMC8032904 DOI: 10.3389/fncel.2021.652593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are increasingly recognized as vital players in the pathology of a variety of neurodegenerative conditions including Alzheimer’s (AD) and Parkinson’s (PD) disease. While microglia have a protective role in the brain, their dysfunction can lead to neuroinflammation and contributes to disease progression. Also, a growing body of literature highlights the seven phosphoinositides, or PIPs, as key players in the regulation of microglial-mediated neuroinflammation. These small signaling lipids are phosphorylated derivates of phosphatidylinositol, are enriched in the brain, and have well-established roles in both homeostasis and disease.Disrupted PIP levels and signaling has been detected in a variety of dementias. Moreover, many known AD disease modifiers identified via genetic studies are expressed in microglia and are involved in phospholipid metabolism. One of these, the enzyme PLCγ2 that hydrolyzes the PIP species PI(4,5)P2, displays altered expression in AD and PD and is currently being investigated as a potential therapeutic target.Perhaps unsurprisingly, neurodegenerative conditions exhibiting PIP dyshomeostasis also tend to show alterations in aspects of microglial function regulated by these lipids. In particular, phosphoinositides regulate the activities of proteins and enzymes required for endocytosis, toll-like receptor signaling, purinergic signaling, chemotaxis, and migration, all of which are affected in a variety of neurodegenerative conditions. These functions are crucial to allow microglia to adequately survey the brain and respond appropriately to invading pathogens and other abnormalities, including misfolded proteins. AD and PD therapies are being developed to target many of the above pathways, and although not yet investigated, simultaneous PIP manipulation might enhance the beneficial effects observed. Currently, only limited therapeutics are available for dementia, and although these show some benefits for symptom severity and progression, they are far from curative. Given the importance of microglia and PIPs in dementia development, this review summarizes current research and asks whether we can exploit this information to design more targeted, or perhaps combined, dementia therapeutics. More work is needed to fully characterize the pathways discussed in this review, but given the strength of the current literature, insights in this area could be invaluable for the future of neurodegenerative disease research.
Collapse
Affiliation(s)
| | - Emily Maguire
- UK Dementia Research Institute at Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
50
|
Dhakal S, Wyant CE, George HE, Morgan SE, Rangachari V. Prion-like C-Terminal Domain of TDP-43 and α-Synuclein Interact Synergistically to Generate Neurotoxic Hybrid Fibrils. J Mol Biol 2021; 433:166953. [PMID: 33771571 DOI: 10.1016/j.jmb.2021.166953] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/04/2021] [Accepted: 03/17/2021] [Indexed: 12/17/2022]
Abstract
Aberrant aggregation and amyloid formation of tar DNA binding protein (TDP-43) and α-synuclein (αS) underlie frontotemporal dementia (FTD) and Parkinson's disease (PD), respectively. Amyloid inclusions of TDP-43 and αS are also commonly co-observed in amyotrophic lateral sclerosis (ALS), dementia with Lewy bodies (DLB) and Alzheimer disease (AD). Emerging evidence from cellular and animal models show colocalization of the TDP-43 and αS aggregates, raising the possibility of direct interactions and co-aggregation between the two proteins. In this report, we set out to answer this question by investigating the interactions between αS and prion-like pathogenic C-terminal domain of TDP-43 (TDP-43 PrLD). PrLD is an aggregation-prone fragment generated both by alternative splicing as well as aberrant proteolytic cleavage of full length TDP-43. Our results indicate that two proteins interact in a synergistic manner to augment each other's aggregation towards hybrid fibrils. While monomers, oligomers and sonicated fibrils of αS seed TDP-43 PrLD monomers, TDP-43 PrLD fibrils failed to seed αS monomers indicating selectivity in interactions. Furthermore, αS modulates liquid droplets formed by TDP-43 PrLD and RNA to promote insoluble amyloid aggregates. Importantly, the cross-seeded hybrid aggregates show greater cytotoxicity as compared to the individual homotypic aggregates suggesting that the interactions between the two proteins have a discernable impact on cellular functions. Together, these results bring forth insights into TDP-43 PrLD - αS interactions that could help explain clinical and pathological presentations in patients with co-morbidities involving the two proteins.
Collapse
Affiliation(s)
- Shailendra Dhakal
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Courtney E Wyant
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Hannah E George
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Sarah E Morgan
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA.
| |
Collapse
|