1
|
Arnold MR, Cohn GM, Oxe KC, Elliott SN, Moore C, Zhou AM, Laraia PV, Shekoohi S, Brownell D, Sears RC, Woltjer RL, Meshul CK, Witt SN, Larsen DH, Unni VK. Alpha-synuclein regulates nucleolar DNA double-strand break repair in melanoma. SCIENCE ADVANCES 2025; 11:eadq2519. [PMID: 40203113 PMCID: PMC11980859 DOI: 10.1126/sciadv.adq2519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Although an increased risk of the skin cancer melanoma in people with Parkinson's disease (PD) has been shown in multiple studies, the mechanisms involved are poorly understood, but increased expression of the PD-associated protein alpha-synuclein (αSyn) in melanoma cells may be important. Our previous work suggests that αSyn can facilitate DNA double-strand break (DSB) repair, promoting genomic stability. We now show that αSyn is preferentially enriched within the nucleolus in melanoma, where it colocalizes with DNA damage markers and DSBs. Inducing DSBs specifically within nucleolar ribosomal DNA (rDNA) increases αSyn levels near sites of damage. αSyn knockout increases DNA damage within the nucleolus at baseline, after specific rDNA DSB induction, and prolongs the rate of recovery from this induced damage. αSyn is important downstream of ataxia-telangiectasia-mutated signaling to facilitate MDC1-mediated 53BP1 recruitment to DSBs, reducing micronuclei formation and promoting cellular proliferation, migration, and invasion.
Collapse
Affiliation(s)
- Moriah R. Arnold
- Medical Scientist Training Program, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Gabriel M. Cohn
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Kezia Catharina Oxe
- Danish Cancer Institute, Nucleolar Stress and Disease Group, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Somarr N. Elliott
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Cynthia Moore
- Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR, USA
| | - Allison May Zhou
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | | | - Sahar Shekoohi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Dillon Brownell
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Randall L. Woltjer
- Layton Aging & Alzheimer’s Disease Research Center and Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Charles K. Meshul
- Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR, USA
- Departments of Behavioral Neuroscience and Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Stephan N. Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Dorthe H. Larsen
- Danish Cancer Institute, Nucleolar Stress and Disease Group, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Vivek K. Unni
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
- OHSU Parkinson Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
2
|
Jimenez-Capdeville ME, Chi-Ahumada E, García-Ortega F, Castanedo-Cazares JP, Norman R, Rodríguez-Leyva I. Nuclear Alpha-Synuclein in Parkinson's Disease and the Malignant Transformation in Melanoma. Neurol Res Int 2025; 2025:1119424. [PMID: 39816956 PMCID: PMC11729518 DOI: 10.1155/nri/1119424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/11/2024] [Indexed: 01/18/2025] Open
Abstract
Background: Alpha-synuclein (ASyn), a marker of Parkinson's disease (PD) and other neurodegenerative processes, plays pivotal roles in neuronal nuclei and synapses. ASyn and its phosphorylated form at Serine 129 (p-ASyn) are involved in DNA protection and repair, processes altered in aging, neurodegeneration, and cancer. Objective: To analyze the localization of p-ASyn in skin biopsies of PD patients and melanoma. Methods: Biopsies from 26 PD patients, 20 melanoma patients, and 31 control subjects were probed and analyzed with a p-ASyn antibody by immunohistochemistry and immunofluorescence. Nuclear positivity was quantified by image analysis. Results: Peripheral nerve endings from healthy subjects show little p-ASyn immunopositivity but notable axonal presence in PD. Control subjects show immunopositivity to p-ASyn along all epidermic strata and scarce presence in their cytoplasm. In contrast, its nuclear presence in PD is weaker, with a higher cytoplasmic and intercellular presence. Nuclear p-ASyn in melanoma varied from similar to control skin in early stage melanoma to a higher rate of empty nuclei in the intermediate stage and total absence of nuclear p-ASyn in severe cases. Interpretation: These findings support the nuclear localization of p-ASyn in skin cells and show that its presence decreases PD and almost disappears in the malignant transformation of melanocytes, redistributing to the cytoplasm and intercellular spaces. This confirms the association between PD and melanoma, providing crucial insights into the role of p-ASyn in both diseases. Trial Registration: ClinicalTrials.gov identifier: NCT01380899.
Collapse
Affiliation(s)
- María E. Jimenez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
| | - Francisco García-Ortega
- Coordinación Académica Región Altiplano, Universidad Autonóma de San Luis Potosi, Matehuala, San Luis Potosí, Mexico
| | | | - Robert Norman
- Center for Geriatric Dermatology, Integrative Dermatology and Euro-Dermatology, Tampa, Florida, USA
| | - Ildefonso Rodríguez-Leyva
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosi, Mexico
- Departamento de Neurología, Hospital “Ignacio Morones Prieto”, San Luis Potosí, Mexico
| |
Collapse
|
3
|
Tian Z, Zhang Y, Xu J, Yang Q, Hu D, Feng J, Gai C. Primary cilia in Parkinson's disease: summative roles in signaling pathways, genes, defective mitochondrial function, and substantia nigra dopaminergic neurons. Front Aging Neurosci 2024; 16:1451655. [PMID: 39364348 PMCID: PMC11447156 DOI: 10.3389/fnagi.2024.1451655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Primary cilia (PC) are microtubules-based, independent antennal-like sensory organelles, that are seen in most vertebrate cells of different types, including astrocytes and neurons. They send signals to cells to control many physiological and cellular processes by detecting changes in the extracellular environment. Parkinson's disease (PD), a neurodegenerative disease that progresses over time, is primarily caused by a gradual degradation of the dopaminergic pathway in the striatum nigra, which results in a large loss of neurons in the substantia nigra compact (SNpc) and a depletion of dopamine (DA). PD samples have abnormalities in the structure and function of PC. The alterations contribute to the cause, development, and recovery of PD via influencing signaling pathways (SHH, Wnt, Notch-1, α-syn, and TGFβ), genes (MYH10 and LRRK2), defective mitochondrial function, and substantia nigra dopaminergic neurons. Thus, restoring the normal structure and physiological function of PC and neurons in the brain are effective treatment for PD. This review summarizes the function of PC in neurodegenerative diseases and explores the pathological mechanisms caused by PC alterations in PD, in order to provide references and ideas for future research.
Collapse
Affiliation(s)
- Zijiao Tian
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yixin Zhang
- College of Acupuncture and Massage, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Xu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qianwen Yang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Die Hu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Feng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Gai
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Duranti E, Villa C. Insights into Dysregulated Neurological Biomarkers in Cancer. Cancers (Basel) 2024; 16:2680. [PMID: 39123408 PMCID: PMC11312413 DOI: 10.3390/cancers16152680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The link between neurodegenerative diseases (NDs) and cancer has generated greater interest in biomedical research, with decades of global studies investigating neurodegenerative biomarkers in cancer to better understand possible connections. Tau, amyloid-β, α-synuclein, SOD1, TDP-43, and other proteins associated with nervous system diseases have also been identified in various types of solid and malignant tumors, suggesting a potential overlap in pathological processes. In this review, we aim to provide an overview of current evidence on the role of these proteins in cancer, specifically examining their effects on cell proliferation, apoptosis, chemoresistance, and tumor progression. Additionally, we discuss the diagnostic and therapeutic implications of this interconnection, emphasizing the importance of further research to completely comprehend the clinical implications of these proteins in tumors. Finally, we explore the challenges and opportunities in targeting these proteins for the development of new targeted anticancer therapies, providing insight into how to integrate knowledge of NDs in oncology research.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
5
|
Amagai R, Otomo R, Yoshioka S, Nagano H, Hashimoto N, Sakakibara R, Tanaka T, Okado-Matsumoto A. C-terminal truncation is a prominent post-translational modification of human erythrocyte α-synuclein. J Biochem 2024; 175:649-658. [PMID: 38308089 DOI: 10.1093/jb/mvae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
α-Synuclein (α-Syn) is a protein related to synucleinopathies with high expression in the central nervous system and erythrocytes which are a major source of peripheral α-Syn. Recent reports have suggested the presence of α-Syn within extracellular vesicles (EVs) derived from erythrocytes, potentially contributing to the pathogenesis of synucleinopathies. While Lewy bodies, intracellular inclusions containing aggregated α-Syn, are prominently observed within the brain, their occurrence in peripheral neurons implies the dissemination of synucleinopathy pathology throughout the body via the propagation of α-Syn. In this study, we found erythrocytes and circulating EVs obtained from plasma contained α-Syn, which was separated into four major forms using high-resolution clear native-PAGE and isoelectric focusing. Notably, erythrocyte α-Syn was classified into full-length and C-terminal truncated forms, with truncation observed between Y133 and Q134 as determined by LC-MS/MS analysis. Our finding revealed that C-terminally truncated α-Syn, which was previously reported to exist solely within the brain, was also present in erythrocytes and circulating EVs obtained from plasma.
Collapse
Affiliation(s)
- Ryosuke Amagai
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Riki Otomo
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Sakura Yoshioka
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Hidekazu Nagano
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Naoko Hashimoto
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ryuji Sakakibara
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Chiba 285-8741, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ayako Okado-Matsumoto
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
6
|
Surguchov A, Surguchev AA. Association between Parkinson's Disease and Cancer: New Findings and Possible Mediators. Int J Mol Sci 2024; 25:3899. [PMID: 38612708 PMCID: PMC11011322 DOI: 10.3390/ijms25073899] [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: 12/20/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Epidemiological evidence points to an inverse association between Parkinson's disease (PD) and almost all cancers except melanoma, for which this association is positive. The results of multiple studies have demonstrated that patients with PD are at reduced risk for the majority of neoplasms. Several potential biological explanations exist for the inverse relationship between cancer and PD. Recent results identified several PD-associated proteins and factors mediating cancer development and cancer-associated factors affecting PD. Accumulating data point to the role of genetic traits, members of the synuclein family, neurotrophic factors, the ubiquitin-proteasome system, circulating melatonin, and transcription factors as mediators. Here, we present recent data about shared pathogenetic factors and mediators that might be involved in the association between these two diseases. We discuss how these factors, individually or in combination, may be involved in pathology, serve as links between PD and cancer, and affect the prevalence of these disorders. Identification of these factors and investigation of their mechanisms of action would lead to the discovery of new targets for the treatment of both diseases.
Collapse
Affiliation(s)
- Andrei Surguchov
- Department of Neurology, Kansas University Medical Center, Kansas City, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Alexei A Surguchev
- Department of Surgery, Section of Otolaryngology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Hansen ML, Ambjørn M, Harndahl MN, Benned-Jensen T, Fog K, Bjerregaard-Andersen K, Sotty F. Characterization of pSer129-αSyn Pathology and Neurofilament Light-Chain Release across In Vivo, Ex Vivo, and In Vitro Models of Pre-Formed-Fibril-Induced αSyn Aggregation. Cells 2024; 13:253. [PMID: 38334646 PMCID: PMC10854598 DOI: 10.3390/cells13030253] [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: 12/04/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Protein aggregation is a predominant feature of many neurodegenerative diseases, including synucleinopathies, which are characterized by cellular inclusions containing α-Synuclein (αSyn) phosphorylated at serine 129 (pSer129). In the present study, we characterized the development of αSyn pre-formed fibril (PFF)-induced pSer129-αSyn pathology in F28tg mice overexpressing human wild-type αSyn, as well as in ex vivo organotypic cultures and in vitro primary cultures from the same mouse model. Concurrently, we collected cerebrospinal fluid (CSF) from mice and conditioned media from ex vivo and in vitro cultures and quantified the levels of neurofilament light chain (NFL), a biomarker of neurodegeneration. We found that the intra-striatal injection of PFFs induces the progressive spread of pSer129-αSyn pathology and microglial activation in vivo, as well as modest increases in NFL levels in the CSF. Similarly, PFF-induced αSyn pathology occurs progressively in ex vivo organotypic slice cultures and is accompanied by significant increases in NFL release into the media. Using in vitro primary hippocampal cultures, we further confirmed that pSer129-αSyn pathology and NFL release occur in a manner that correlates with the fibril dose and the level of the αSyn protein. Overall, we demonstrate that αSyn pathology is associated with NFL release across preclinical models of seeded αSyn aggregation and that the pharmacological inhibition of αSyn aggregation in vitro also significantly reduces NFL release.
Collapse
Affiliation(s)
- Maja L. Hansen
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Malene Ambjørn
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Mikkel N. Harndahl
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.N.H.); (K.B.-A.)
| | - Tau Benned-Jensen
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | - Karina Fog
- Neuroscience, Molecular and Cellular Pharmacology, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark; (M.L.H.); (M.A.); (K.F.)
| | | | - Florence Sotty
- Neuroscience, Histology and Pathology Models, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| |
Collapse
|
8
|
Arnold MR, Cohn GM, Oxe KC, Elliott SN, Moore C, Laraia PV, Shekoohi S, Brownell D, Meshul CK, Witt SN, Larsen DH, Unni VK. Alpha-synuclein regulates nucleolar DNA double-strand break repair in melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.575526. [PMID: 38260370 PMCID: PMC10802588 DOI: 10.1101/2024.01.13.575526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Although an increased risk of the skin cancer melanoma in people with Parkinson's Disease (PD) has been shown in multiple studies, the mechanisms involved are poorly understood, but increased expression of the PD-associated protein alpha-synuclein (αSyn) in melanoma cells may be important. Our previous work suggests that αSyn can facilitate DNA double-strand break (DSB) repair, promoting genomic stability. We now show that αSyn is preferentially enriched within the nucleolus in the SK-MEL28 melanoma cell line, where it colocalizes with DNA damage markers and DSBs. Inducing DSBs specifically within nucleolar ribosomal DNA (rDNA) increases αSyn levels near sites of damage. αSyn knockout increases DNA damage within the nucleolus at baseline, after specific rDNA DSB induction, and prolongs the rate of recovery from this induced damage. αSyn is important downstream of ATM signaling to facilitate 53BP1 recruitment to DSBs, reducing micronuclei formation and promoting cellular proliferation, migration, and invasion.
Collapse
Affiliation(s)
- Moriah R. Arnold
- Medical Scientist Training Program, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Gabriel M. Cohn
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Kezia Catharina Oxe
- Danish Cancer Institute, Nucleolar Stress and Disease Group, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Somarr N. Elliott
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Cynthia Moore
- Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR, USA
| | | | - Sahar Shekoohi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Dillon Brownell
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Charles K. Meshul
- Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR, USA
- Departments of Behavioral Neuroscience and Pathology, Oregon Health and Science University, Portland, OR, USA
| | - Stephan N. Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Dorthe H. Larsen
- Danish Cancer Institute, Nucleolar Stress and Disease Group, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Vivek K. Unni
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
- OHSU Parkinson’s Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
9
|
Bétemps D, Arsac JN, Nicot S, Canal D, Tlili H, Belondrade M, Morignat E, Verchère J, Gaillard D, Bruyère-Ostells L, Mayran C, Lakhdar L, Bougard D, Baron T. Protease-Sensitive and -Resistant Forms of Human and Murine Alpha-Synucleins in Distinct Brain Regions of Transgenic Mice (M83) Expressing the Human Mutated A53T Protein. Biomolecules 2023; 13:1788. [PMID: 38136658 PMCID: PMC10741842 DOI: 10.3390/biom13121788] [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: 09/27/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Human neurodegenerative diseases associated with the misfolding of the alpha-synuclein (aS) protein (synucleinopathies) are similar to prion diseases to the extent that lesions are spread by similar molecular mechanisms. In a transgenic mouse model (M83) overexpressing a mutated (A53T) form of human aS, we had previously found that Protein Misfolding Cyclic Amplification (PMCA) triggered the aggregation of aS, which is associated with a high resistance to the proteinase K (PK) digestion of both human and murine aS, a major hallmark of the disease-associated prion protein. In addition, PMCA was also able to trigger the aggregation of murine aS in C57Bl/6 mouse brains after seeding with sick M83 mouse brains. Here, we show that intracerebral inoculations of M83 mice with C57Bl/6-PMCA samples strikingly shortens the incubation period before the typical paralysis that develops in this transgenic model, demonstrating the pathogenicity of PMCA-aggregated murine aS. In the hind brain regions of these sick M83 mice containing lesions with an accumulation of aS phosphorylated at serine 129, aS also showed a high PK resistance in the N-terminal part of the protein. In contrast to M83 mice, old APPxM83 mice co-expressing human mutated amyloid precursor and presenilin 1 proteins were seen to have an aggregation of aS, especially in the cerebral cortex, hippocampus and striatum, which also contained the highest load of aS phosphorylated at serine 129. This was proven by three techniques: a Western blot analysis of PK-resistant aS; an ELISA detection of aS aggregates; or the identification of aggregates of aS using immunohistochemical analyses of cytoplasmic/neuritic aS deposits. The results obtained with the D37A6 antibody suggest a higher involvement of murine aS in APPxM83 mice than in M83 mice. Our study used novel tools for the molecular study of synucleinopathies, which highlight similarities with the molecular mechanisms involved in prion diseases.
Collapse
Affiliation(s)
- Dominique Bétemps
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Jean-Noël Arsac
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Simon Nicot
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Dominique Canal
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Habiba Tlili
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Maxime Belondrade
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Eric Morignat
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Jérémy Verchère
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Damien Gaillard
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Lilian Bruyère-Ostells
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Charly Mayran
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Latifa Lakhdar
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| | - Daisy Bougard
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Inserm, Etablissement Français Du Sang, 34493 Montpellier, France; (S.N.); (M.B.); (L.B.-O.); (C.M.); (D.B.)
| | - Thierry Baron
- ANSES (French Agency for Food, Environmental and Occupational Health & Safety), University of Lyon, 69364 Lyon, France; (D.B.); (J.-N.A.); (D.C.); (H.T.); (E.M.); (J.V.); (D.G.); (L.L.)
| |
Collapse
|
10
|
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Santos-García D, Martínez-Valbuena I, Agúndez JAG. Alpha-Synuclein in Peripheral Tissues as a Possible Marker for Neurological Diseases and Other Medical Conditions. Biomolecules 2023; 13:1263. [PMID: 37627328 PMCID: PMC10452242 DOI: 10.3390/biom13081263] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The possible usefulness of alpha-synuclein (aSyn) determinations in peripheral tissues (blood cells, salivary gland biopsies, olfactory mucosa, digestive tract, skin) and in biological fluids, except for cerebrospinal fluid (serum, plasma, saliva, feces, urine), as a marker of several diseases, has been the subject of numerous publications. This narrative review summarizes data from studies trying to determine the role of total, oligomeric, and phosphorylated aSyn determinations as a marker of various diseases, especially PD and other alpha-synucleinopathies. In summary, the results of studies addressing the determinations of aSyn in its different forms in peripheral tissues (especially in platelets, skin, and digestive tract, but also salivary glands and olfactory mucosa), in combination with other potential biomarkers, could be a useful tool to discriminate PD from controls and from other causes of parkinsonisms, including synucleinopathies.
Collapse
Affiliation(s)
| | | | - Elena García-Martín
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| | - Diego Santos-García
- Department of Neurology, CHUAC—Complejo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain;
| | - Iván Martínez-Valbuena
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 2S8, Canada;
| | - José A. G. Agúndez
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, 10071 Cáceres, Spain; (E.G.-M.); (J.A.G.A.)
| |
Collapse
|
11
|
Hou TZ, Yang HM, Cheng YZ, Gu L, Zhang JN, Zhang H. The Parkinson's disease-associated protein α-synuclein inhibits hepatoma by exosome delivery. Mol Carcinog 2023; 62:1163-1175. [PMID: 37144864 DOI: 10.1002/mc.23553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/31/2023] [Accepted: 04/22/2023] [Indexed: 05/06/2023]
Abstract
Numerous epidemiological studies suggest a link between Parkinson's disease (PD) and cancer. However, their relevant pathogenesis is not clear. In the present study, we investigated the potential role of exosome-delivered α-synuclein (α-syn) in the regulation between PD and liver cancer. We cultured hepatocellular carcinoma (HCC) cells with exosomes derived from conditioned medium of the PD cellular model, and injected exosomes enriched with α-syn into the striatum of a liver cancer rat model. We found that α-syn-contained exosomes from the rotenone-induced cellular model of PD suppressed the growth, migration, and invasion of HCC cells. Integrin αVβ5 in exosomes from the rotenone-induced PD model was higher than that in the control, resulting in more α-syn-contained exosomes being taken up by HCC cells. Consistently, in vivo experiments with rat models also confirmed exosome-delivered α-syn inhibited liver cancer. These findings illustrate the important role of PD-associated protein α-syn inhibiting hepatoma by exosome delivery, suggesting a new mechanism underlying the link between these two diseases and therapeutics of liver cancer.
Collapse
Affiliation(s)
- Tian-Zhong Hou
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Yun-Zhong Cheng
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
12
|
Silva JL, Foguel D, Ferreira VF, Vieira TCRG, Marques MA, Ferretti GDS, Outeiro TF, Cordeiro Y, de Oliveira GAP. Targeting Biomolecular Condensation and Protein Aggregation against Cancer. Chem Rev 2023. [PMID: 37379327 DOI: 10.1021/acs.chemrev.3c00131] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Biomolecular condensates, membrane-less entities arising from liquid-liquid phase separation, hold dichotomous roles in health and disease. Alongside their physiological functions, these condensates can transition to a solid phase, producing amyloid-like structures implicated in degenerative diseases and cancer. This review thoroughly examines the dual nature of biomolecular condensates, spotlighting their role in cancer, particularly concerning the p53 tumor suppressor. Given that over half of the malignant tumors possess mutations in the TP53 gene, this topic carries profound implications for future cancer treatment strategies. Notably, p53 not only misfolds but also forms biomolecular condensates and aggregates analogous to other protein-based amyloids, thus significantly influencing cancer progression through loss-of-function, negative dominance, and gain-of-function pathways. The exact molecular mechanisms underpinning the gain-of-function in mutant p53 remain elusive. However, cofactors like nucleic acids and glycosaminoglycans are known to be critical players in this intersection between diseases. Importantly, we reveal that molecules capable of inhibiting mutant p53 aggregation can curtail tumor proliferation and migration. Hence, targeting phase transitions to solid-like amorphous and amyloid-like states of mutant p53 offers a promising direction for innovative cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Jerson L Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Debora Foguel
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Vitor F Ferreira
- Faculty of Pharmacy, Fluminense Federal University (UFF), Rio de Janeiro, RJ 21941-902, Brazil
| | - Tuane C R G Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Mayra A Marques
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Giulia D S Ferretti
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center, 37075 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, U.K
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
13
|
Fujii T, Nagamori S, Wiriyasermkul P, Zheng S, Yago A, Shimizu T, Tabuchi Y, Okumura T, Fujii T, Takeshima H, Sakai H. Parkinson's disease-associated ATP13A2/PARK9 functions as a lysosomal H +,K +-ATPase. Nat Commun 2023; 14:2174. [PMID: 37080960 PMCID: PMC10119128 DOI: 10.1038/s41467-023-37815-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Mutations in the human ATP13A2 (PARK9), a lysosomal ATPase, cause Kufor-Rakeb Syndrome, an early-onset form of Parkinson's disease (PD). Here, we demonstrate that ATP13A2 functions as a lysosomal H+,K+-ATPase. The K+-dependent ATPase activity and the lysosomal K+-transport activity of ATP13A2 are inhibited by an inhibitor of sarco/endoplasmic reticulum Ca2+-ATPase, thapsigargin, and K+-competitive inhibitors of gastric H+,K+-ATPase, such as vonoprazan and SCH28080. Interestingly, these H+,K+-ATPase inhibitors cause lysosomal alkalinization and α-synuclein accumulation, which are pathological hallmarks of PD. Furthermore, PD-associated mutants of ATP13A2 show abnormal expression and function. Our results suggest that the H+/K+-transporting function of ATP13A2 contributes to acidification and α-synuclein degradation in lysosomes.
Collapse
Affiliation(s)
- Takuto Fujii
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Shushi Nagamori
- Center for SI Medical Research and Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Pattama Wiriyasermkul
- Center for SI Medical Research and Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Shizhou Zheng
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Asaka Yago
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Takahiro Shimizu
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Hideki Sakai
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
14
|
Zhou LX, Zheng H, Tian Y, Luo KF, Ma SJ, Wu ZW, Tang P, Jiang J, Wang MH. SNCA inhibits epithelial-mesenchymal transition and correlates to favorable prognosis of breast cancer. Carcinogenesis 2022; 43:1071-1082. [PMID: 36179220 DOI: 10.1093/carcin/bgac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/03/2022] [Accepted: 09/29/2022] [Indexed: 02/04/2023] Open
Abstract
Alpha-synuclein (SNCA) is a pathological hallmark of Parkinson's disease, known to be involved in cancer occurrence and development; however, its specific effects in breast cancer remain unknown. Data from 150 patients with breast cancer were retrieved from tissue microarray and analyzed for SNCA protein level using immunohistochemistry. Functional enrichment analysis was performed to investigate the potential role of SNCA in breast cancer. SNCA-mediated inhibition of epithelial-mesenchymal transition (EMT) was confirmed with western blotting. The effects of SNCA on invasion and migration were evaluated using transwell and wound-healing experiments. Furthermore, the potential influence of SNCA expression level on drug sensitivity and tumor infiltration by immune cells was analyzed using the public databases. SNCA is lowly expressed in breast cancer tissues. Besides, in vitro and in vivo experiments, SNCA overexpression blocked EMT and metastasis, and the knockdown of SNCA resulted in the opposite effect. A mouse model of metastasis verified the restriction of metastatic ability in vivo. Further analysis revealed that SNCA enhances sensitivity to commonly used anti-breast tumor drugs and immune cell infiltration. SNCA blocks EMT and metastasis in breast cancer and its expression levels could be useful in predicting the chemosensitivity and evaluating the immune microenvironment in breast cancer.
Collapse
Affiliation(s)
- Lin-Xi Zhou
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400038, China
| | - Yuan Tian
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
- Department of Emergency Surgery, Linyi People's Hospital, Linyi 276000, China
| | - Ke-Fei Luo
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Shu-Juan Ma
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Zi-Wei Wu
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Peng Tang
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Jun Jiang
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Ming-Hao Wang
- Breast Disease Center, Southwest Hospital, Army Medical University, Chongqing 400038, China
| |
Collapse
|
15
|
Bianchini M, Giambelluca M, Scavuzzo MC, Di Franco G, Guadagni S, Palmeri M, Furbetta N, Gianardi D, Costa A, Gentiluomo M, Gaeta R, Pollina LE, Falcone A, Vivaldi C, Di Candio G, Biagioni F, Busceti CL, Soldani P, Puglisi-Allegra S, Morelli L, Fornai F. In Pancreatic Adenocarcinoma Alpha-Synuclein Increases and Marks Peri-Neural Infiltration. Int J Mol Sci 2022; 23:3775. [PMID: 35409135 PMCID: PMC8999122 DOI: 10.3390/ijms23073775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
α-Synuclein (α-syn) is a protein involved in neuronal degeneration. However, the family of synucleins has recently been demonstrated to be involved in the mechanisms of oncogenesis by selectively accelerating cellular processes leading to cancer. Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers, with a specifically high neurotropism. The molecular bases of this biological behavior are currently poorly understood. Here, α-synuclein was analyzed concerning the protein expression in PDAC and the potential association with PDAC neurotropism. Tumor (PDAC) and extra-tumor (extra-PDAC) samples from 20 patients affected by PDAC following pancreatic resections were collected at the General Surgery Unit, University of Pisa. All patients were affected by moderately or poorly differentiated PDAC. The amount of α-syn was compared between tumor and extra-tumor specimen (sampled from non-affected neighboring pancreatic areas) by using in situ immuno-staining with peroxidase anti-α-syn immunohistochemistry, α-syn detection by using Western blotting, and electron microscopy by using α-syn-conjugated immuno-gold particles. All the methods consistently indicate that each PDAC sample possesses a higher amount of α-syn compared with extra-PDAC tissue. Moreover, the expression of α-syn was much higher in those PDAC samples from tumors with perineural infiltration compared with tumors without perineural infiltration.
Collapse
Affiliation(s)
- Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Maria Giambelluca
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.G.); (M.C.S.); (P.S.)
| | - Maria Concetta Scavuzzo
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.G.); (M.C.S.); (P.S.)
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Simone Guadagni
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Niccolò Furbetta
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Aurelio Costa
- General Surgery Unit, ASL Toscana Nord Ovest Pontedera Hospital, 56025 Pontedera, Italy;
| | | | - Raffaele Gaeta
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, 56124 Pisa, Italy; (R.G.); (L.E.P.)
| | - Luca Emanuele Pollina
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, 56124 Pisa, Italy; (R.G.); (L.E.P.)
| | - Alfredo Falcone
- Division of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (A.F.); (C.V.)
| | - Caterina Vivaldi
- Division of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (A.F.); (C.V.)
| | - Giulio Di Candio
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
| | - Francesca Biagioni
- IRCCS Neuromed-Istituto Neurologico Mediterraneo, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (S.P.-A.)
| | - Carla Letizia Busceti
- IRCCS Neuromed-Istituto Neurologico Mediterraneo, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (S.P.-A.)
| | - Paola Soldani
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.G.); (M.C.S.); (P.S.)
| | - Stefano Puglisi-Allegra
- IRCCS Neuromed-Istituto Neurologico Mediterraneo, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (S.P.-A.)
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.B.); (G.D.F.); (S.G.); (M.P.); (N.F.); (D.G.); (G.D.C.)
- EndoCAS (Center for Computer Assisted Surgery), University of Pisa, 56124 Pisa, Italy
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (M.G.); (M.C.S.); (P.S.)
- IRCCS Neuromed-Istituto Neurologico Mediterraneo, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (S.P.-A.)
| |
Collapse
|
16
|
Ryskalin L, Biagioni F, Morucci G, Busceti CL, Frati A, Puglisi-Allegra S, Ferrucci M, Fornai F. Spreading of Alpha Synuclein from Glioblastoma Cells towards Astrocytes Correlates with Stem-like Properties. Cancers (Basel) 2022; 14:cancers14061417. [PMID: 35326570 PMCID: PMC8946011 DOI: 10.3390/cancers14061417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The present study questions whether cells from glioblastoma multiforme (GBM), which overexpress α-synuclein (α-syn), may alter neighboring non-tumoral astrocyte cell lines. The occurrence of α-syn in GBM correlates with the expression of the stem cell marker nestin. When astrocytes are co-cultured with GBM cells in a trans-well apparatus the occurrence of α-syn and nestin rises remarkably. The increase in α-syn in co-cultured astrocytes is more pronounced at the plasma membrane, which mimics the placement of α-syn in GBM cells. When the mTOR inhibitor rapamycin is administered, GBM-induced expression of α-syn and nestin within co-cultured astrocytes is occluded, and morphological alterations are reverted. In the presence of rapamycin the sub-cellular placement of α-syn is modified being allocated within whorls and vacuoles instead of the plasma membrane. The effects induced by rapamycin occur both in baseline GBM cells and within astrocytes primed by co-cultured GBM cells. Abstract Evidence has been recently provided showing that, in baseline conditions, GBM cells feature high levels of α-syn which are way in excess compared with α-syn levels measured within control astrocytes. These findings are consistent along various techniques. In fact, they are replicated by using antibody-based protein detection, such as immuno-fluorescence, immuno-peroxidase, immunoblotting and ultrastructural stoichiometry as well as by measuring α-syn transcript levels at RT-PCR. The present manuscript further questions whether such a high amount of α-syn may be induced within astrocytes, which are co-cultured with GBM cells in a trans-well system. In astrocytes co-cultured with GBM cells, α-syn overexpression is documented. Such an increase is concomitant with increased expression of the stem cell marker nestin, along with GBM-like shifting in cell morphology. This concerns general cell morphology, subcellular compartments and profuse convolutions at the plasma membrane. Transmission electron microscopy (TEM) allows us to assess the authentic amount and sub-cellular compartmentalization of α-syn and nestin within baseline GBM cells and the amount, which is induced within co-cultured astrocytes, as well as the shifting of ultrastructure, which is reminiscent of GBM cells. These phenomena are mitigated by rapamycin administration, which reverts nestin- and α-syn-related overexpression and phenotypic shifting within co-cultured astrocytes towards baseline conditions of naïve astrocytes. The present study indicates that: (i) α-syn increases in astrocyte co-cultured with GBM cells; (ii) α-syn increases in astrocytes along with the stem cell marker nestin; (iii) α-syn increases along with a GBM-like shift of cell morphology; (iv) all these changes are replicated in different GBM cell lines and are reverted by the mTOR inhibitor rapamycin. The present findings indicate that α-syn does occur in high amount within GBM cells and may transmit to neighboring astrocytes as much as a stem cell phenotype. This suggests a mode of tumor progression for GBM cells, which may transform, rather than merely substitute, surrounding tissue; such a phenomenon is sensitive to mTOR inhibition.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (G.M.); (M.F.)
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (G.M.); (M.F.)
| | - Carla L. Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135 Roma, Italy
| | - Stefano Puglisi-Allegra
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (G.M.); (M.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (G.M.); (M.F.)
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.); (S.P.-A.)
- Correspondence: ; Tel.: +39-050-2218601
| |
Collapse
|
17
|
Occurrence of Total and Proteinase K-Resistant Alpha-Synuclein in Glioblastoma Cells Depends on mTOR Activity. Cancers (Basel) 2022; 14:cancers14061382. [PMID: 35326535 PMCID: PMC8946689 DOI: 10.3390/cancers14061382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 01/18/2023] Open
Abstract
Simple Summary The accumulation of alpha-synuclein (α-syn) is considered a pathological hallmark of the neurodegenerative disorders known as synucleinopathies. The clearance of α-syn depends on autophagy activity, which is inhibited by the mechanistic target of rapamycin (mTOR). Thus, it is likely that α-syn accumulation may occur whenever mTOR is overactive and autophagy is suppressed. In fact, the lack of effective autophagy increases the amount of α-syn and may produce protein aggregation. Therefore, in the present study, we questioned whether cells from glioblastoma multiforme (GBM), a lethal brain neoplasm, wherein mTOR is upregulated and autophagy is suppressed, may overexpress α-syn. In fact, a large quantity of α-syn is measured in GBM cells compared with astrocytes, which includes proteinase K-resistant α-syn. Rapamycin, while inhibiting mTOR activity, significantly reduces the amount of α-syn and allocates α-syn within autophagy-like vacuoles. Abstract Alpha-synuclein (α-syn) is a protein considered to be detrimental in a number of degenerative disorders (synucleinopathies) of which α-syn aggregates are considered a pathological hallmark. The clearance of α-syn strongly depends on autophagy, which can be stimulated by inhibiting the mechanistic target of rapamycin (mTOR). Thus, the overexpression of mTOR and severe autophagy suppression may produce α-syn accumulation, including the proteinase K-resistant protein isoform. Glioblastoma multiforme (GBM) is a lethal brain tumor that features mTOR overexpression and severe autophagy inhibition. Cell pathology in GBM is reminiscent of a fast, progressive degenerative disorder. Therefore, the present work questions whether, as is analogous to neurons during degenerative disorders, an overexpression of α-syn occurs within GBM cells. A high amount of α-syn was documented in GBM cells via real-time PCR (RT-PCR), Western blotting, immunohistochemistry, immuno-fluorescence, and ultrastructural stoichiometry, compared with the amount of β- and γ-synucleins and compared with the amount of α-syn counted within astrocytes. The present study indicates that (i) α-syn is overexpressed in GBM cells, (ii) α-syn expression includes a proteinase-K resistant isoform, (iii) α-syn is dispersed from autophagy-like vacuoles to the cytosol, (iv) α-syn overexpression and cytosol dispersion are mitigated by rapamycin, and (v) the α-syn-related GBM-like phenotype is mitigated by silencing the SNCA gene.
Collapse
|
18
|
Dent SE, King DP, Osterberg VR, Adams EK, Mackiewicz MR, Weissman TA, Unni VK. Phosphorylation of the aggregate-forming protein alpha-synuclein on serine-129 inhibits its DNA-bending properties. J Biol Chem 2021; 298:101552. [PMID: 34973339 PMCID: PMC8800120 DOI: 10.1016/j.jbc.2021.101552] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
Alpha-synuclein (aSyn) is a vertebrate protein, normally found within the presynaptic nerve terminal and nucleus, which is known to form somatic and neuritic aggregates in certain neurodegenerative diseases. Disease-associated aggregates of aSyn are heavily phosphorylated at serine-129 (pSyn), while normal aSyn protein is not. Within the nucleus, aSyn can directly bind DNA, but the mechanism of binding and the potential modulatory roles of phosphorylation are poorly understood. Here we demonstrate using a combination of electrophoretic mobility shift assay and atomic force microscopy approaches that both aSyn and pSyn can bind DNA within the major groove, in a DNA length-dependent manner and with little specificity for DNA sequence. Our data are consistent with a model in which multiple aSyn molecules bind a single 300 base pair (bp) DNA molecule in such a way that stabilizes the DNA in a bent conformation. We propose that serine-129 phosphorylation decreases the ability of aSyn to both bind and bend DNA, as aSyn binds 304 bp circular DNA forced into a bent shape, but pSyn does not. Two aSyn paralogs, beta- and gamma-synuclein, also interact with DNA differently than aSyn, and do not stabilize similar DNA conformations. Our work suggests that reductions in aSyn's ability to bind and bend DNA induced by serine-129 phosphorylation may be important for modulating aSyn's known roles in DNA metabolism, including the regulation of transcription and DNA repair.
Collapse
Affiliation(s)
- Sydney E Dent
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Dennisha P King
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Valerie R Osterberg
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Eleanor K Adams
- Department of Chemistry, Portland State University, Portland, Oregon, 97239, USA
| | - Marilyn R Mackiewicz
- Department of Chemistry, Portland State University, Portland, Oregon, 97239, USA
| | - Tamily A Weissman
- Department of Biology, Lewis & Clark College, Portland, Oregon, 97219, USA
| | - Vivek K Unni
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon, 97239, USA; OHSU Parkinson Center, Oregon Health & Science University, Portland, Oregon, 97239, USA.
| |
Collapse
|
19
|
Dean DN, Lee JC. Linking Parkinson's Disease and Melanoma: Interplay Between α-Synuclein and Pmel17 Amyloid Formation. Mov Disord 2021; 36:1489-1498. [PMID: 34021920 DOI: 10.1002/mds.28655] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with the death of dopaminergic neurons within the substantia nigra of the brain. Melanoma is a cancer of melanocytes, pigmented cells that give rise to skin tone, hair, and eye color. Although these two diseases fundamentally differ, with PD leading to cell degeneration and melanoma leading to cell proliferation, epidemiological evidence has revealed a reciprocal relationship where patients with PD are more susceptible to melanoma and patients with melanoma are more susceptible to PD. The hallmark pathology observed in PD brains is intracellular inclusions, of which the primary component is proteinaceous α-synuclein (α-syn) amyloid fibrils. α-Syn also has been detected in cultured melanoma cells and tissues derived from patients with melanoma, where an inverse correlation exists between α-syn expression and pigmentation. Although this has led to the prevailing hypothesis that α-syn inhibits enzymes involved in melanin biosynthesis, we recently reported an alternative hypothesis in which α-syn interacts with and modulates the aggregation of Pmel17, a functional amyloid that serves as a scaffold for melanin biosynthesis. In this perspective, we review the literature describing the epidemiological and molecular connections between PD and melanoma, presenting both the prevailing hypothesis and our amyloid-centric hypothesis. We offer our views of the essential questions that remain unanswered to motivate future investigations. Understanding the behavior of α-syn in melanoma could not only provide novel approaches for treating melanoma but also could reveal insights into the role of α-syn in PD. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dexter N Dean
- Laboratory of Protein Conformation and Dynamics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer C Lee
- Laboratory of Protein Conformation and Dynamics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Molecular Characteristics of Amyloid Precursor Protein (APP) and Its Effects in Cancer. Int J Mol Sci 2021; 22:ijms22094999. [PMID: 34066808 PMCID: PMC8125876 DOI: 10.3390/ijms22094999] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
Amyloid precursor protein (APP) is a type 1 transmembrane glycoprotein, and its homologs amyloid precursor-like protein 1 (APLP1) and amyloid precursor-like protein 2 (APLP2) are highly conserved in mammals. APP and APLP are known to be intimately involved in the pathogenesis and progression of Alzheimer's disease and to play important roles in neuronal homeostasis and development and neural transmission. APP and APLP are also expressed in non-neuronal tissues and are overexpressed in cancer cells. Furthermore, research indicates they are involved in several cancers. In this review, we examine the biological characteristics of APP-related family members and their roles in cancer.
Collapse
|
21
|
Laferrière F, He X, Zinghirino F, Doudnikoff E, Faggiani E, Meissner WG, Bezard E, De Giorgi F, Ichas F. Overexpression of α-Synuclein by Oligodendrocytes in Transgenic Mice Does Not Recapitulate the Fibrillar Aggregation Seen in Multiple System Atrophy. Cells 2020; 9:E2371. [PMID: 33138150 PMCID: PMC7693764 DOI: 10.3390/cells9112371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 11/17/2022] Open
Abstract
The synucleinopathy underlying multiple system atrophy (MSA) is characterized by the presence of abundant amyloid inclusions containing fibrillar α-synuclein (α-syn) aggregates in the brains of the patients and is associated with an extensive neurodegeneration. In contrast to Parkinson's disease (PD) where the pathological α-syn aggregates are almost exclusively neuronal, the α-syn inclusions in MSA are principally observed in oligodendrocytes (OLs) where they form glial cytoplasmic inclusions (GCIs). This is intriguing because differentiated OLs express low levels of α-syn, yet pathogenic amyloid α-syn seeds require significant amounts of α-syn monomers to feed their fibrillar growth and to eventually cause the buildup of cytopathological inclusions. One of the transgenic mouse models of this disease is based on the targeted overexpression of human α-syn in OLs using the PLP promoter. In these mice, the histopathological images showing a rapid emergence of S129-phosphorylated α-syn inside OLs are considered as equivalent to GCIs. Instead, we report here that they correspond to the accumulation of phosphorylated α-syn monomers/oligomers and not to the appearance of the distinctive fibrillar α-syn aggregates that are present in the brains of MSA or PD patients. In spite of a propensity to co-sediment with myelin sheath contaminants, the phosphorylated forms found in the brains of the transgenic animals are soluble (>80%). In clear contrast, the phosphorylated species present in the brains of MSA and PD patients are insoluble fibrils (>95%). Using primary cultures of OLs from PLP-αSyn mice we observed a variable association of S129-phosphorylated α-syn with the cytoplasmic compartment, the nucleus and with membrane domains suggesting that OLs functionally accommodate the phospho-α-syn deriving from experimental overexpression. Yet and while not taking place spontaneously, fibrillization can be seeded in these primary cultures by challenging the OLs with α-syn preformed fibrils (PFFs). This indicates that a targeted overexpression of α-syn does not model GCIs in mice but that it can provide a basis for seeding aggregation using PFFs. This approach could help establishing a link between α-syn aggregation and the development of a clinical phenotype in these transgenic animals.
Collapse
Affiliation(s)
- Florent Laferrière
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Xin He
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
- Department of Neurology, Sheng Jing Hospital of China Medical University, Shenyang 110004, China
| | - Federica Zinghirino
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
- Dipartimento di Scienze Biomediche e Biotecnologiche, BIOMETEC, Università degli Studi di Catania, 95123 Catania, Italy
| | - Evelyne Doudnikoff
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Emilie Faggiani
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Wassilios G. Meissner
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
- Service de Neurologie, CRMR Atrophie Multisystématisée, CHU Bordeaux, 33000 Bordeaux, France
| | - Erwan Bezard
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francesca De Giorgi
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
- INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, U-1084, Université de Poitiers, 86000 Poitiers, France
| | - François Ichas
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33076 Bordeaux, France; (F.L.); (X.H.); (F.Z.); (E.D.); (E.F.); (W.G.M.); (E.B.)
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
- INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, U-1084, Université de Poitiers, 86000 Poitiers, France
| |
Collapse
|
22
|
Ejma M, Madetko N, Brzecka A, Guranski K, Alster P, Misiuk-Hojło M, Somasundaram SG, Kirkland CE, Aliev G. The Links between Parkinson's Disease and Cancer. Biomedicines 2020; 8:biomedicines8100416. [PMID: 33066407 PMCID: PMC7602272 DOI: 10.3390/biomedicines8100416] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Epidemiologic studies indicate a decreased incidence of most cancer types in Parkinson’s disease (PD) patients. However, some neoplasms are associated with a higher risk of occurrence in PD patients. Both pathologies share some common biological pathways. Although the etiologies of PD and cancer are multifactorial, some factors associated with PD, such as α-synuclein aggregation; mutations of PINK1, PARKIN, and DJ-1; mitochondrial dysfunction; and oxidative stress can also be involved in cancer proliferation or cancer suppression. The main protein associated with PD, i.e., α-synuclein, can be involved in some types of neoplastic formations. On the other hand, however, its downregulation has been found in the other cancers. PINK1 can act as oncogenic or a tumor suppressor. PARKIN dysfunction may lead to some cancers’ growth, and its expression may be associated with some tumors’ suppression. DJ-1 mutation is involved in PD pathogenesis, but its increased expression was found in some neoplasms, such as melanoma or breast, lung, colorectal, uterine, hepatocellular, and nasopharyngeal cancers. Both mitochondrial dysfunction and oxidative stress are involved in PD and cancer development. The aim of this review is to summarize the possible associations between PD and carcinogenesis.
Collapse
Affiliation(s)
- Maria Ejma
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Natalia Madetko
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, Grabiszyńska 105, 53-439 Wroclaw, Poland;
| | - Konstanty Guranski
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Piotr Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warszawa, Poland;
| | - Marta Misiuk-Hojło
- Department of Ophthalmology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia
- Research Institute of Human Morphology, Russian Academy of Medical Science, Street Tsyurupa 3, 117418 Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432 Moscow Region, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
- Correspondence: or ; Tel.: +1-210-442-8625 or +1-440-263-7461
| |
Collapse
|
23
|
Vijayan B, Raj V, Nandakumar S, Kishore A, Thekkuveettil A. Spermine protects alpha-synuclein expressing dopaminergic neurons from manganese-induced degeneration. Cell Biol Toxicol 2018; 35:147-159. [PMID: 30673990 DOI: 10.1007/s10565-018-09449-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/16/2018] [Indexed: 12/14/2022]
Abstract
Manganese exposure is among the many environmental risk factors linked to the progression of neurodegenerative diseases, such as manganese-induced parkinsonism. In animal models, chronic exposure to manganese causes loss of cell viability, neurodegeneration, and functional deficits. Polyamines, such as spermine, have been shown to rescue animals from age-induced neurodegeneration in an autophagy-dependent manner; nonetheless, it is not understood whether polyamines can prevent manganese-induced toxicity. In this study, we used two model systems, the Caenorhabditis elegans UA44 strain and SK-MEL-28 cells, both expressing the protein alpha-synuclein (α-syn) to determine whether spermine could ameliorate manganese-induced toxicity. Manganese caused a substantial reduction in the viability of SK-MEL-28 cells and hastened neurodegeneration in the UA44 strain. Spermine protected both the SK-MEL-28 cells and the UA44 strain from manganese-induced toxicity. Spermine also reduced the age-associated neurodegeneration observed in the UA44 strain compared with a control strain without α-syn expression and led to improved avoidance behavior in a functional assay. Treatment with berenil, an inhibitor of polyamine catabolism, which leads to increased intracellular polyamine levels, also showed similar cellular protection against manganese toxicity. While both translation blocker cycloheximide and autophagy blocker chloroquine caused a reduction in the cytoprotective effect of spermine, transcription blocker actinomycin D had no effect. This study provides new insights on the effect of spermine in preventing manganese-induced toxicity, which is most likely via translational regulation of several candidate genes, including those of autophagy. Thus, our results indicate that polyamines positively influence neuronal health, even when exposed to high levels of manganese and α-syn, and supplementing polyamines through diet might delay the onset of diseases involving degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Bejoy Vijayan
- Comprehensive Care Centre for Movement Disorders, Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Vishnu Raj
- Division of Molecular Medicine, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Swapna Nandakumar
- Comprehensive Care Centre for Movement Disorders, Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Asha Kishore
- Comprehensive Care Centre for Movement Disorders, Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Anoopkumar Thekkuveettil
- Division of Molecular Medicine, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
24
|
Autophagy enhancement is rendered ineffective in presence of α-synuclein in melanoma cells. J Cell Commun Signal 2017; 11:381-394. [PMID: 28748508 DOI: 10.1007/s12079-017-0402-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022] Open
Abstract
Increased cellular concentration of α-synuclein (α-syn) predisposes it to misfolding and aggregation that in turn impair the degradation pathways. This poses a limitation to the use of overexpression models for studies on α-syn clearance by autophagy, which is widely investigated for its therapeutic potential. This limitation can be overcome with the use of endogenous models. In this study, SK-MEL-28, a melanoma cell model with endogenous α-syn expression, was employed to study α-syn clearance through autophagy. We demonstrated the dual localization of α-syn to nucleus and cytoplasm that varied in response to changes in cellular environment. Autophagy inhibition and exposure to dopamine favored cytoplasmic localization of α-syn, while autophagy induction favored increased localization to the nucleus. The inhibitory effect of dopamine on autophagy was heightened in presence of α-syn. Additionally, because α-syn had a regulatory effect on autophagy, cells showed an increased resistance to autophagy induction in presence of α-syn. This resistance prevented effective induction of autophagy even under conditions of prolonged autophagy inhibition. These results highlight alternate physiological roles of α-syn, particularly in non-neuronal cells. Because autophagy enhancement could reverse neither the increase in α-syn levels nor the autophagy inhibition, there arises a need to evaluate the efficacy of autophagy-based therapeutic strategies.
Collapse
|
25
|
Turriani E, Lázaro DF, Ryazanov S, Leonov A, Giese A, Schön M, Schön MP, Griesinger C, Outeiro TF, Arndt-Jovin DJ, Becker D. Treatment with diphenyl-pyrazole compound anle138b/c reveals that α-synuclein protects melanoma cells from autophagic cell death. Proc Natl Acad Sci U S A 2017; 114:E4971-E4977. [PMID: 28584093 PMCID: PMC5488931 DOI: 10.1073/pnas.1700200114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recent epidemiological and clinical studies have reported a significantly increased risk for melanoma in people with Parkinson's disease. Because no evidence could be obtained that genetic factors are the reason for the association between these two diseases, we hypothesized that of the three major Parkinson's disease-related proteins-α-synuclein, LRRK2, and Parkin-α-synuclein might be a major link. Our data, presented here, demonstrate that α-synuclein promotes the survival of primary and metastatic melanoma cells, which is the exact opposite of the effect that α-synuclein has on dopaminergic neurons, where its accumulation causes neuronal dysfunction and death. Because this detrimental effect of α-synuclein on neurons can be rescued by the small molecule anle138b, we explored its effect on melanoma cells. We found that treatment with anle138b leads to massive melanoma cell death due to a major dysregulation of autophagy, suggesting that α-synuclein is highly beneficial to advanced melanoma because it ensures that autophagy is maintained at a homeostatic level that promotes and ensures the cell's survival.
Collapse
Affiliation(s)
- Elisa Turriani
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Diana F Lázaro
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Sergey Ryazanov
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Georg-August-University Göttingen, 37073 Göttingen, Germany
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Andrei Leonov
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Georg-August-University Göttingen, 37073 Göttingen, Germany
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Margarete Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Christian Griesinger
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Georg-August-University Göttingen, 37073 Göttingen, Germany
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Georg-August-University Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Donna J Arndt-Jovin
- Laboratory of Cellular Dynamics, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Dorothea Becker
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany;
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| |
Collapse
|
26
|
Rodriguez-Leyva I, Chi-Ahumada E, Mejía M, Castanedo-Cazares JP, Eng W, Saikaly SK, Carrizales J, Levine TD, Norman RA, Jimenez-Capdeville ME. The Presence of Alpha-Synuclein in Skin from Melanoma and Patients with Parkinson's Disease. Mov Disord Clin Pract 2017; 4:724-732. [PMID: 30363411 DOI: 10.1002/mdc3.12494] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 12/31/2022] Open
Abstract
Background The misfolding and prion-like propagation of the protein α-synuclein (α-syn) is the leading molecular signature in Parkinson's disease (PD). There is a significant coincidence of PD and melanoma that may suggest a shared pathophysiology. This study compared the presence of α-syn in neural crest-derived tissues, such as nevi, melanoma, skin tags, and skin biopsies from patients with PD and healthy controls. Methods Biopsies from participants with PD were obtained from patients from a tertiary referral center for dermatology and neurology in Mexico and a private dermatopathology center in Florida between January 2015 and March 2016. Biopsies from 7 patients with melanoma, 15 with nevi, 9 with skin tags, 8 with PD, and 9 skin biopsies from healthy volunteers were analyzed for immunohistochemical determination of α-syn and tyrosinase. All analyses were performed by pathologists who were blinded with respect to the clinical diagnosis. Results In healthy controls, positive α-syn status was restricted to scattered cells in the basal layer of the epidermis and accounted for 1 ± 0.8% of the analyzed area. In patients with PD, there was increased staining for α-syn PD (3.3 ± 2.3%), with a higher percentage of positive cells in nevi (7.7 ± 5.5%) and melanoma (13.6 ± 3.5%). There was no increased staining in skin tags compared with healthy controls. Conclusion Patients with PD and melanoma have increased staining for α-syn in their skin. The authors propose that neurons and melanocytes, both derived from neuroectodermal cells, may share protein synthesis and regulation pathways that become dysfunctional in PD and melanoma.
Collapse
Affiliation(s)
| | - Erika Chi-Ahumada
- Departamento de Bioquímica Facultad de Medicina Universidad Autónoma de San Luis Potosí San Luis Potosí México
| | - Manuel Mejía
- Departamento de Bioquímica Facultad de Medicina Universidad Autónoma de San Luis Potosí San Luis Potosí México
| | | | - William Eng
- University of Central Florida College of Medicine Orlando Florida USA
| | - Sami K Saikaly
- University of Central Florida College of Medicine Orlando Florida USA
| | - Juan Carrizales
- Departamento de Bioquímica Facultad de Medicina Universidad Autónoma de San Luis Potosí San Luis Potosí México
| | | | - Robert A Norman
- University of Central Florida College of Medicine Orlando Florida USA
| | - Maria E Jimenez-Capdeville
- Departamento de Bioquímica Facultad de Medicina Universidad Autónoma de San Luis Potosí San Luis Potosí México
| |
Collapse
|
27
|
Nanoparticle standards for immuno-based quantitation of α-synuclein oligomers in diagnostics of Parkinson's disease and other synucleinopathies. Clin Chim Acta 2017; 466:152-159. [DOI: 10.1016/j.cca.2017.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/22/2016] [Accepted: 01/10/2017] [Indexed: 11/17/2022]
|
28
|
Oueslati A. Implication of Alpha-Synuclein Phosphorylation at S129 in Synucleinopathies: What Have We Learned in the Last Decade? JOURNAL OF PARKINSONS DISEASE 2017; 6:39-51. [PMID: 27003784 PMCID: PMC4927808 DOI: 10.3233/jpd-160779] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abnormal accumulation of proteinaceous intraneuronal inclusions called Lewy bodies (LBs) is the neurpathological hallmark of Parkinson’s disease (PD) and related synucleinopathies. These inclusions are mainly constituted of a presynaptic protein, α-synuclein (α-syn). Over the past decade, growing amounts of studies reported an aberrant accumulation of phosphorylated α-syn at the residue S129 (pS129) in the brain of patients suffering from PD, as well as in transgenic animal models of synucleinopathies. Whereas only a small fraction of α-syn (<4%) is phosphorylated in healthy brains, a dramatic accumulation of pS129 (>90%) has been observed within LBs, suggesting that this post-translational modification may play an important role in the regulation of α-syn aggregation, LBs formation and neuronal degeneration. However, whether phosphorylation at S129 suppresses or enhances α-syn aggregation and toxicity in vivo remains a subject of active debate. The answer to this question has important implications for understanding the role of phosphorylation in the pathogenesis of synucleinopathies and determining if targeting kinases or phosphatases could be a viable therapeutic strategy for the treatment of these devastating neurological disorders. In the present review, we explore recent findings from in vitro, cell-based assays and in vivo studies describing the potential implications of pS129 in the regulation of α-syn physiological functions, as well as its implication in synucleinopathies pathogenesis and diagnosis.
Collapse
Affiliation(s)
- Abid Oueslati
- Correspondence to: Abid Oueslati, Centre de Recherche du CHU de Québec-Université Laval, Axe Neuroscience et Départe-ment de Médecine Moléculaire de l’Université Laval, Québec G1V4G2, Canada. Tel.: +1 4185254444/Ext 49119; Fax: +1 4186542125; E-mail:
| |
Collapse
|
29
|
Surguchov A. Parkinson's Disease: Assay of Phosphorylated α-Synuclein in Skin Biopsy for Early Diagnosis and Association with Melanoma. Brain Sci 2016; 6:E17. [PMID: 27240409 PMCID: PMC4931494 DOI: 10.3390/brainsci6020017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is a degenerative disorder of the central nervous system, in which a small naturally unfolded protein α-synuclein plays an essential role. α-Synuclein belongs to a synuclein family comprising three members: α, β, and γ-synucleins associated with neurodegenerative and neoplastic diseases and involved in development. Several studies revealed that α-synuclein is present not only in the brain, but also in the skin and other peripheral tissues. This finding open a new approach to PD diagnosis based on the assay of α-synuclein from a biological sample of a living patient. Furthermore, PD is associated with an increased risk of skin melanoma. An important posttranslational modification of α-synuclein is phosphorylation at serine-129, which may convert the protein into pathological species both in PD and melanoma. Thus, analysis of phosphorylated α-synuclein might be an important diagnostic test for both diseases providing additional information about the mechanism of pathology.
Collapse
Affiliation(s)
- Andrei Surguchov
- Department of Neurology, Kansas University Medical Center, Kansas City, 3901, Rainbow Boulevard, Kansas City, KS 66160, USA.
| |
Collapse
|
30
|
Feng DD, Cai W, Chen X. The associations between Parkinson's disease and cancer: the plot thickens. Transl Neurodegener 2015; 4:20. [PMID: 26504519 PMCID: PMC4620601 DOI: 10.1186/s40035-015-0043-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/07/2015] [Indexed: 01/10/2023] Open
Abstract
Epidemiological studies support a general inverse association between the risk of cancer development and Parkinson’s disease (PD). In recent years however, increasing amount of eclectic evidence points to a positive association between PD and cancers through different temporal analyses and ethnic groups. This positive association has been supported by several common genetic mutations in SNCA, PARK2, PARK8, ATM, p53, PTEN, and MC1R resulting in cellular changes such as mitochondrial dysfunction, aberrant protein aggregation, and cell cycle dysregulation. Here, we review the epidemiological and biological advances of the past decade in the association between PD and cancers to offer insight on the recent and sometimes contradictory findings.
Collapse
Affiliation(s)
- Danielle D Feng
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Waijiao Cai
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA.,Key Laboratory of Cellular and Molecular Biology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
31
|
Sasaki A, Arawaka S, Sato H, Kato T. Sensitive western blotting for detection of endogenous Ser129-phosphorylated α-synuclein in intracellular and extracellular spaces. Sci Rep 2015; 5:14211. [PMID: 26381815 PMCID: PMC4585644 DOI: 10.1038/srep14211] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/20/2015] [Indexed: 11/29/2022] Open
Abstract
α-Synuclein deposited in Lewy bodies, a pathological hallmark of Parkinson’s disease (PD), is highly phosphorylated at serine 129 (Ser129). In contrast, there is very little Ser129-phosphorylated α-synuclein in the normal brains. This difference suggests that Ser129-phosphorylation is involved in neurodegenerative processes of PD. However, the role of this modification remains unclear. One limiting factor for relevant biochemical analyses is that it is difficult to detect endogenous Ser129-phosphoryated α-synuclein by western blotting, because α-synuclein monomers detached from the transferred membrane during incubation. Here, we reported that combination fixation of the transferred membrane with 4% paraformaldehyde and 0.01 ~ 0.1% glutaraldehyde produced an approximately 10-fold increase in the sensitivity for Ser129-phosphorylated α-synuclein monomers, allowing detection of endogenous proteins even in conditioned medium, human cerebrospinal fluid, and extracts from cell lines and human brain. This method may enable more detailed biochemical analyses for α-synuclein transmission between intra and extracellular spaces under physiological and pathological conditions.
Collapse
Affiliation(s)
- Asuka Sasaki
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata University Faculty of Medicine. 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Shigeki Arawaka
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata University Faculty of Medicine. 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Hiroyasu Sato
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata University Faculty of Medicine. 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Takeo Kato
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata University Faculty of Medicine. 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| |
Collapse
|
32
|
Bétemps D, Verchère J, Mougenot AL, Lachmann I, Morignat E, Antier E, Lakhdar L, Legastelois S, Baron T. Detection of Disease-associated α-synuclein by Enhanced ELISA in the Brain of Transgenic Mice Overexpressing Human A53T Mutated α-synuclein. J Vis Exp 2015:e52752. [PMID: 26068223 DOI: 10.3791/52752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
In addition to established methods like Western blot, new methods are needed to quickly and easily quantify disease-associated α-synuclein (αS(D)) in experimental models of synucleopathies. A transgenic mouse line (M83) over-expressing the human A53T αS and spontaneously developing a dramatic clinical phenotype between eight and 22 months of age, characterized by symptoms including weight loss, prostration, and severe motor impairment, was used in this study. For molecular analyses of αS(D) (disease-associated αS) in these mice, an ELISA was designed to specifically quantify αS(D) in sick mice. Analysis of the central nervous system in this mouse model showed the presence of αS(D) mainly in the caudal brain regions and the spinal cord. There were no differences in αS(D) distribution between different experimental conditions leading to clinical disease, i.e., in uninoculated and normally aging transgenic mice and in mice inoculated with brain extracts from sick mice. The specific detection of αS(D) immunoreactivity using an antibody against Ser129 phosphorylated αS by ELISA essentially correlated with that obtained by Western blot and immunohistochemistry. Unexpectedly, similar results were observed with several other antibodies against the C-terminal part of αS. The propagation of αS(D), suggesting the involvement of a "prion-like" mechanism, can thus be easily monitored and quantified in this mouse model using an ELISA approach.
Collapse
Affiliation(s)
- Dominique Bétemps
- Neurodegenerative Diseases Unit, ANSES - French Agency for Food, Environmental and Occupational Health & Safety
| | - Jérémy Verchère
- Neurodegenerative Diseases Unit, ANSES - French Agency for Food, Environmental and Occupational Health & Safety
| | | | | | - Eric Morignat
- Epidemiology Unit, ANSES - French Agency for Food, Environmental and Occupational Health & Safety
| | - Emilie Antier
- Experimental Facilities, ANSES - French Agency for Food, Environmental and Occupational Health & Safety
| | - Latifa Lakhdar
- Experimental Facilities, ANSES - French Agency for Food, Environmental and Occupational Health & Safety
| | | | - Thierry Baron
- Neurodegenerative Diseases Unit, ANSES - French Agency for Food, Environmental and Occupational Health & Safety;
| |
Collapse
|
33
|
Guardia-Laguarta C, Area-Gomez E, Schon EA, Przedborski S. A new role for α-synuclein in Parkinson's disease: Alteration of ER-mitochondrial communication. Mov Disord 2015; 30:1026-33. [DOI: 10.1002/mds.26239] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 03/10/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022] Open
Affiliation(s)
| | - Estela Area-Gomez
- Department of Neurology; Columbia University Medical Center; New York NY USA
| | - Eric A. Schon
- Department of Neurology; Columbia University Medical Center; New York NY USA
- Department of Genetics and Development; Columbia University Medical Center; New York NY USA
| | - Serge Przedborski
- Department of Pathology and Cell Biology; Columbia University Medical Center; New York NY USA
| |
Collapse
|
34
|
Burai R, Ait-Bouziad N, Chiki A, Lashuel HA. Elucidating the Role of Site-Specific Nitration of α-Synuclein in the Pathogenesis of Parkinson's Disease via Protein Semisynthesis and Mutagenesis. J Am Chem Soc 2015; 137:5041-52. [PMID: 25768729 DOI: 10.1021/ja5131726] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and the presence of intraneuronal inclusions consisting of aggregated and post-translationally modified α-synuclein (α-syn). Despite advances in the chemical synthesis of α-syn and other proteins, the generation of site-specifically nitrated synthetic proteins has not been reported. Consequently, it has not been possible to determine the roles of nitration at specific residues in regulating the physiological and pathogenic properties of α-syn. Here we report, for the first time, the site-specific incorporation of 3-nitrotyrosine at different regions of α-syn using native chemical ligation combined with a novel desulfurization strategy. This strategy enabled us to investigate the role of nitration at single or multiple tyrosine residues in regulating α-syn structure, membrane binding, oligomerization, and fibrils formation. We demonstrate that different site-specifically nitrated α-syn species exhibit distinct structural and aggregation properties and exhibit reduced affinity to negatively charged vesicle membranes. We provide evidence that intermolecular interactions between the N- and C-terminal regions of α-syn play critical roles in mediating nitration-induced α-syn oligomerization. For example, when Y39 is not available for nitration (Y39F and Y39/125F), the extent of cross-linking is limited mostly to dimer formation, whereas mutants in which Y39 along with one or multiple C-terminal tyrosines (Y125F, Y133F, Y136F and Y133/136F) can still undergo nitration readily to form higher-order oligomers. Our semisynthetic strategy for generating site-specifically nitrated proteins opens up new possibilities for investigating the role of nitration in regulating protein structure and function in health and disease.
Collapse
Affiliation(s)
- Ritwik Burai
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Nadine Ait-Bouziad
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Anass Chiki
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Bétemps D, Verchère J, Brot S, Morignat E, Bousset L, Gaillard D, Lakhdar L, Melki R, Baron T. Alpha-synuclein spreading in M83 mice brain revealed by detection of pathological α-synuclein by enhanced ELISA. Acta Neuropathol Commun 2014; 2:29. [PMID: 24624994 PMCID: PMC4007641 DOI: 10.1186/2051-5960-2-29] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/20/2014] [Indexed: 11/16/2022] Open
Abstract
Background The accumulation of misfolded proteins appears as a fundamental pathogenic process in human neurodegenerative diseases. In the case of synucleinopathies such as Parkinson’s disease (PD) or dementia with Lewy bodies (DLB), the intraneuronal deposition of aggregated alpha-synuclein (αS) is a major characteristic of the disease, but the molecular basis distinguishing the disease-associated protein (αSD) from its normal counterpart remains poorly understood. However, recent research suggests that a prion-like mechanism could be involved in the inter-cellular and inter-molecular propagation of aggregation of the protein within the nervous system. Results Our data confirm our previous observations of disease acceleration in a transgenic mouse line (M83) overexpressing a mutated (A53T) form of human αS, following inoculation of either brain extracts from sick M83 mice or fibrillar recombinant αS. A similar phenomenon is observed following a “second passage” in the M83 mouse model, including after stereotactic inoculations into the hippocampus or cerebellum. For further molecular analyses of αSD, we designed an ELISA test that identifies αSD specifically in sick mice and in the brain regions targeted by the pathological process in this mouse model. αSD distribution, mainly in the caudal brain regions and spinal cord, overall appears remarkably uniform, whatever the conditions of experimental challenge. In addition to specific detection of αSD immunoreactivity using an antibody against Ser129 phosphorylated αS, similar results were observed in ELISA with several other antibodies against the C-terminal part of αS, including an antibody against non phosphorylated αS. This also indicated consistent immunoreactivity of the murine αS protein specifically in the affected brain regions of sick mice. Conclusions Prion-like behaviour in propagation of the disease-associated αS was confirmed with the M83 transgenic mouse model, that could be followed by an ELISA test. The ELISA data question their possible relationship with the conformational differences between the disease-associated αS and its normal counterpart.
Collapse
|
36
|
|