1
|
Li S, Liu Y, Lu S, Xu J, Liu X, Yang D, Yang Y, Hou L, Li N. A crazy trio in Parkinson's disease: metabolism alteration, α-synuclein aggregation, and oxidative stress. Mol Cell Biochem 2025; 480:139-157. [PMID: 38625515 DOI: 10.1007/s11010-024-04985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
Parkinson's disease (PD) is an aging-associated neurodegenerative disorder, characterized by the progressive loss of dopaminergic neurons in the pars compacta of the substantia nigra and the presence of Lewy bodies containing α-synuclein within these neurons. Oligomeric α-synuclein exerts neurotoxic effects through mitochondrial dysfunction, glial cell inflammatory response, lysosomal dysfunction and so on. α-synuclein aggregation, often accompanied by oxidative stress, is generally considered to be a key factor in PD pathology. At present, emerging evidences suggest that metabolism alteration is closely associated with α-synuclein aggregation and PD progression, and improvement of key molecules in metabolism might be potentially beneficial in PD treatment. In this review, we highlight the tripartite relationship among metabolic changes, α-synuclein aggregation, and oxidative stress in PD, and offer updated insights into the treatments of PD, aiming to deepen our understanding of PD pathogenesis and explore new therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sheng Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yanbing Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sen Lu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiayi Xu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaokun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Di Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ning Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
2
|
Liu X, Liu X, Liu Y, Yang B, Li Y, Li F, Qian K, Liu X, Xiao L, Cui G, Xu C. Exploratory Analysis of the Association Between Plasma Ceramide Alterations and Cognitive Dysfunction in Parkinson's Disease. CNS Neurosci Ther 2024; 30:e70082. [PMID: 39428566 PMCID: PMC11491299 DOI: 10.1111/cns.70082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/31/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE Prior research has underscored the importance of sphingolipid metabolism in Parkinson's disease (PD) pathogenesis. Our objective was to explore the associations between plasma ceramide levels and PD patients with cognitive dysfunction (PD-CD). METHODS We enrolled two study populations from Eastern China and the Parkinson's Progression Markers Initiative (PPMI), comprising 290 (100 HCs, 160 PDs, and 30 MSAs) and 429 (125 HCs and 304 PDs) participants, respectively. The plasma levels of ceramides (Cer 16:0, Cer 18:0, Cer 24:0, and Cer 24:1) were tested via HPLC-MS/MS analysis. RESULTS Compared with those in the HC group, the plasma levels of Cer 18:0, Cer 24:1, Cer 16:0/Cer 24:0, Cer 18:0/Cer 24:0, and Cer 24:1/Cer 24:0 were higher in both the PD and MSA groups. Significant differences in the plasma levels of Cer 16:0/Cer 24:0, Cer 18:0/Cer 24:0, and Cer 24:1/Cer 24:0 were observed among the PD-NC (PD with normal cognition), PD-MCI (PD with mild cognitive impairment), and PDD (PD dementia) groups, with the PDD group exhibiting the highest levels. PD patients with higher baseline levels of plasma ceramides (specifically, Cer 18:0, Cer 16:0/Cer 24:0, Cer 18:0/Cer 24:0, and Cer 24:1/Cer 24:0) demonstrated accelerated cognitive decline compared with individuals who had lower baseline plasma ceramide levels during the 5-year follow-up period. A biomarker panel including Cer 18:0/Cer 24:0 and Cer 24:1/Cer 24:0 could effectively differentiate PD-CD from PD-NC with notable diagnostic accuracy. CONCLUSIONS Our results indicate that plasma ceramide levels could potentially be used as diagnostic biomarkers for PD-CD.
Collapse
Affiliation(s)
- Xu Liu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of NeurologyThe Second Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Xuanjing Liu
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yuning Liu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Bo Yang
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yangdanyu Li
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Fujia Li
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Kun Qian
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Xuesong Liu
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Lishun Xiao
- Department of Biostatistics, School of Public HealthXuzhou Medical UniversityXuzhouJiangsuChina
| | - Guiyun Cui
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| | - Chuanying Xu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouJiangsuChina
- Department of Neurology, the First Clinical CollegeXuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
3
|
Ribarič S. The Contribution of Type 2 Diabetes to Parkinson's Disease Aetiology. Int J Mol Sci 2024; 25:4358. [PMID: 38673943 PMCID: PMC11050090 DOI: 10.3390/ijms25084358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are chronic disorders that have a significant health impact on a global scale. Epidemiological, preclinical, and clinical research underpins the assumption that insulin resistance and chronic inflammation contribute to the overlapping aetiologies of T2D and PD. This narrative review summarises the recent evidence on the contribution of T2D to the initiation and progression of PD brain pathology. It also briefly discusses the rationale and potential of alternative pharmacological interventions for PD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Kalinichenko LS, Kohl Z, Mühle C, Hassan Z, Hahn A, Schmitt EM, Macht K, Stoyanov L, Moghaddami S, Bilbao R, Eulenburg V, Winkler J, Kornhuber J, Müller CP. Sex-specific pleiotropic changes in emotional behavior and alcohol consumption in human α-synuclein A53T transgenic mice during early adulthood. J Neurochem 2024; 168:269-287. [PMID: 38284431 DOI: 10.1111/jnc.16051] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/15/2023] [Accepted: 01/07/2024] [Indexed: 01/30/2024]
Abstract
Point mutations in the α-synuclein coding gene may lead to the development of Parkinson's disease (PD). PD is often accompanied by other psychiatric conditions, such as anxiety, depression, and drug use disorders, which typically emerge in adulthood. Some of these point mutations, such as SNCA and A30T, have been linked to behavioral effects that are not commonly associated with PD, especially regarding alcohol consumption patterns. In this study, we investigated whether the familial PD point mutation A53T is associated with changes in alcohol consumption behavior and emotional states at ages not yet characterized by α-synuclein accumulation. The affective and alcohol-drinking phenotypes remained unaltered in female PDGF-hA53T-synuclein-transgenic (A53T) mice during both early and late adulthood. Brain region-specific activation of ceramide-producing enzymes, acid sphingomyelinase (ASM), and neutral sphingomyelinase (NSM), known for their neuroprotective properties, was observed during early adulthood but not in late adulthood. In males, the A53T mutation was linked to a reduction in alcohol consumption in both early and late adulthood. However, male A53T mice displayed increased anxiety- and depression-like behaviors during both early and late adulthood. Enhanced ASM activity in the dorsal mesencephalon and ventral hippocampus may potentially contribute to these adverse behavioral effects of the mutation in males during late adulthood. In summary, the A53T gene mutation was associated with diverse changes in emotional states and alcohol consumption behavior long before the onset of PD, and these effects varied by sex. These alterations in behavior may be linked to changes in brain ceramide metabolism.
Collapse
Affiliation(s)
- Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Zacharias Kohl
- Division of Molecular Neurology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Erlangen, Germany
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
| | - Agnes Hahn
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Schmitt
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Macht
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lyubomir Stoyanov
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Schayan Moghaddami
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Roberto Bilbao
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Volker Eulenburg
- Department for Anesthesiology and Intensive Care, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Jürgen Winkler
- Division of Molecular Neurology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Joshi N, Sarhadi TR, Raveendran A, Nagotu S. Sporadic SNCA mutations A18T and A29S exhibit variable effects on protein aggregation, cell viability and oxidative stress. Mol Biol Rep 2023:10.1007/s11033-023-08457-7. [PMID: 37155014 DOI: 10.1007/s11033-023-08457-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND α-synuclein aggregation is the hallmark feature of Parkinson's disease. Both familial and sporadic forms of the disease exhibit this feature. Several mutations have been identified in patients and are associated with the disease pathology. METHODS AND RESULTS We have used site-directed mutagenesis to generate α-synuclein mutant variants tagged with GFP. Fluorescence microscopy, flow cytometry, western blotting, cell viability and oxidative stress analysis were performed to investigate the effect of two less studied α-synuclein variants. In this study we characterized two less studied α-synuclein mutations, A18T and A29S, in the well-established yeast model. Our data shows variable expression, distribution and toxicity of the protein in the mutant variants A18T, A29S, A53T and WT. The cells expressing the double mutant variant A18T/A53T showed the most increase in the aggregation phenotype and also depicted reduced viability suggesting a more substantial effect of this variant. CONCLUSION The outcome of our study highlights the variable localization, aggregation phenotype and toxicity of the studied α-synuclein variants. This underscores the importance of in-depth analysis of every disease-associated mutation which may result in variable cellular phenotype.
Collapse
Affiliation(s)
- Neha Joshi
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Tanveera Rounaque Sarhadi
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Atchaya Raveendran
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Shirisha Nagotu
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
6
|
The Consequences of GBA Deficiency in the Autophagy-Lysosome System in Parkinson's Disease Associated with GBA. Cells 2023; 12:cells12010191. [PMID: 36611984 PMCID: PMC9818455 DOI: 10.3390/cells12010191] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023] Open
Abstract
GBA gene variants were the first genetic risk factor for Parkinson's disease. GBA encodes the lysosomal enzyme glucocerebrosidase (GBA), which is involved in sphingolipid metabolism. GBA exhibits a complex physiological function that includes not only the degradation of its substrate glucosylceramide but also the metabolism of other sphingolipids and additional lipids such as cholesterol, particularly when glucocerebrosidase activity is deficient. In the context of Parkinson's disease associated with GBA, the loss of GBA activity has been associated with the accumulation of α-synuclein species. In recent years, several hypotheses have proposed alternative and complementary pathological mechanisms to explain why lysosomal enzyme mutations lead to α-synuclein accumulation and become important risk factors in Parkinson's disease etiology. Classically, loss of GBA activity has been linked to a dysfunctional autophagy-lysosome system and to a subsequent decrease in autophagy-dependent α-synuclein turnover; however, several other pathological mechanisms underlying GBA-associated parkinsonism have been proposed. This review summarizes and discusses the different hypotheses with a special focus on autophagy-dependent mechanisms, as well as autophagy-independent mechanisms, where the role of other players such as sphingolipids, cholesterol and other GBA-related proteins make important contributions to Parkinson's disease pathogenesis.
Collapse
|
7
|
Annesley SJ, Allan CY, Sanislav O, Evans A, Fisher PR. Dysregulated Gene Expression in Lymphoblasts from Parkinson’s Disease. Proteomes 2022; 10:proteomes10020020. [PMID: 35736800 PMCID: PMC9230639 DOI: 10.3390/proteomes10020020] [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: 04/08/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Parkinson’s disease is the second largest neurodegenerative disease worldwide and is caused by a combination of genetics and environment. It is characterized by the death of neurons in the substantia nigra of the brain but is not solely a disease of the brain, as it affects multiple tissues and organs. Studying Parkinson’s disease in accessible tissues such as skin and blood has increased our understanding of the disease’s pathogenesis. Here, we used lymphoblast cell lines generated from Parkinson’s disease patient and healthy age- and sex-matched control groups and obtained their whole-cell transcriptomes and proteomes. Our analysis revealed, in both the transcriptomes and the proteomes of PD cells, a global downregulation of genes involved in protein synthesis, as well as the upregulation of immune processes and sphingolipid metabolism. In contrast, we discovered an uncoupling of mRNA and protein expression in processes associated with mitochondrial respiration in the form of a general downregulation in associated transcripts and an upregulation in proteins. Complex V was different to the other oxidative phosphorylation complexes in that the levels of its associated transcripts were also lower, but the levels of their encoded polypeptides were not elevated. This may suggest that further layers of regulation specific to Complex V are in play.
Collapse
Affiliation(s)
- Sarah Jane Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC 3086, Australia; (C.Y.A.); (O.S.); (P.R.F.)
- Correspondence: ; Tel.: +61-394791412
| | - Claire Yvonne Allan
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC 3086, Australia; (C.Y.A.); (O.S.); (P.R.F.)
| | - Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC 3086, Australia; (C.Y.A.); (O.S.); (P.R.F.)
| | - Andrew Evans
- Department of Neurology, Royal Melbourne Hospital, Parkville, VIC 3052, Australia;
| | - Paul Robert Fisher
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC 3086, Australia; (C.Y.A.); (O.S.); (P.R.F.)
| |
Collapse
|
8
|
Ozaki K, Irioka T, Uchihara T, Yamada A, Nakamura A, Majima T, Igarashi S, Shintaku H, Yakeishi M, Tsuura Y, Okazaki Y, Ishikawa K, Yokota T. Neuropathology of SCA34 showing widespread oligodendroglial pathology with vacuolar white matter degeneration: a case study. Acta Neuropathol Commun 2021; 9:172. [PMID: 34689836 PMCID: PMC8543940 DOI: 10.1186/s40478-021-01272-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/10/2021] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxia type 34 (SCA34) is an autosomal dominant inherited ataxia due to mutations in ELOVL4, which encodes one of the very long-chain fatty acid elongases. SCA38, another spinocerebellar ataxia, is caused by mutations in ELOVL5, a gene encoding another elongase. However, there have been no previous studies describing the neuropathology of either SCA34 or 38. This report describes the neuropathological findings of an 83-year-old man with SCA34 carrying a pathological ELOVL4 mutation (NM_022726, c.736T>G, p.W246G). Macroscopic findings include atrophies in the pontine base, cerebellum, and cerebral cortices. Microscopically, marked neuronal and pontocerebellar fiber loss was observed in the pontine base. In addition, in the pontine base, accumulation of CD68-positive macrophages laden with periodic acid-Schiff (PAS)-positive material was observed. Many vacuolar lesions were found in the white matter of the cerebral hemispheres and, to a lesser extent, in the brainstem and spinal cord white matter. Immunohistological examination and ultrastructural observations with an electron microscope suggest that these vacuolar lesions are remnants of degenerated oligodendrocytes. Electron microscopy also revealed myelin sheath destruction. Unexpectedly, aggregation of the four-repeat tau was observed in a spatial pattern reminiscent of progressive supranuclear palsy. The tau lesions included glial fibrillary tangles resembling tuft-shaped astrocytes and neurofibrillary tangles and pretangles. This is the first report to illustrate that a heterozygous missense mutation in ELOVL4 leads to neuronal loss accompanied by macrophages laden with PAS-positive material in the pontine base and oligodendroglial degeneration leading to widespread vacuoles in the white matter in SCA34.
Collapse
|
9
|
Rajasekaran S, Peterson PP, Liu Z, Robinson LC, Witt SN. α-Synuclein inhibits Snx3-retromer retrograde trafficking of the conserved membrane-bound proprotein convertase Kex2 in the secretory pathway of Saccharomyces cerevisiae. Hum Mol Genet 2021; 31:705-717. [PMID: 34570221 DOI: 10.1093/hmg/ddab284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
We tested the ability of alpha-synuclein (α-syn) to inhibit Snx3-retromer mediated retrograde trafficking of Kex2 and Ste13 between late endosomes and the trans-Golgi (TGN) using a Saccharomyces cerevisiae model of Parkinson's disease (PD). Kex2 and Ste13 are a conserved, membrane-bound proprotein convertase and dipeptidyl aminopeptidase, respectively, that process pro-α-factor and pro-killer toxin. Each of these proteins contains a cytosolic tail that binds to sorting nexin Snx3. Using a combination of techniques, including fluorescence microscopy, western blotting and a yeast mating assay, we found that α-syn disrupts Snx3-retromer trafficking of Kex2-GFP and GFP-Ste13 from the late endosome to the TGN, resulting in these two proteins transiting to the vacuole by default. Using three α-syn variants (A53T, A30P, and α-synΔC, which lacks residues 101-140), we further found that A53T and α-synΔC, but not A30P, reduce Snx3-retromer trafficking of Kex2-GFP, which is likely to be due to weaker binding of A30P to membranes. Degradation of Kex2 and Ste13 in the vacuole should result in the secretion of unprocessed, inactive forms of α-factor, which will reduce mating efficiency between MATa and MATα cells. We found that wild-type α-syn but not A30P significantly inhibited the secretion of α-factor. Collectively, our results support a model in which the membrane-binding ability of α-syn is necessary to disrupt Snx3-retromer retrograde recycling of these two conserved endopeptidases.
Collapse
Affiliation(s)
- Santhanasabapathy Rajasekaran
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - Patricia P Peterson
- Department of Biological Sciences, The University of New Orleans, New Orleans, LA 70148 USA
| | - Zhengchang Liu
- Department of Biological Sciences, The University of New Orleans, New Orleans, LA 70148 USA
| | - Lucy C Robinson
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| | - Stephan N Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71103 USA
| |
Collapse
|
10
|
Sarchione A, Marchand A, Taymans JM, Chartier-Harlin MC. Alpha-Synuclein and Lipids: The Elephant in the Room? Cells 2021; 10:2452. [PMID: 34572099 PMCID: PMC8467310 DOI: 10.3390/cells10092452] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/17/2022] Open
Abstract
Since the initial identification of alpha-synuclein (α-syn) at the synapse, numerous studies demonstrated that α-syn is a key player in the etiology of Parkinson's disease (PD) and other synucleinopathies. Recent advances underline interactions between α-syn and lipids that also participate in α-syn misfolding and aggregation. In addition, increasing evidence demonstrates that α-syn plays a major role in different steps of synaptic exocytosis. Thus, we reviewed literature showing (1) the interplay among α-syn, lipids, and lipid membranes; (2) advances of α-syn synaptic functions in exocytosis. These data underscore a fundamental role of α-syn/lipid interplay that also contributes to synaptic defects in PD. The importance of lipids in PD is further highlighted by data showing the impact of α-syn on lipid metabolism, modulation of α-syn levels by lipids, as well as the identification of genetic determinants involved in lipid homeostasis associated with α-syn pathologies. While questions still remain, these recent developments open the way to new therapeutic strategies for PD and related disorders including some based on modulating synaptic functions.
Collapse
Affiliation(s)
| | | | | | - Marie-Christine Chartier-Harlin
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172—LilNCog—Lille Neuroscience and Cognition, F-59000 Lille, France; (A.S.); (A.M.); (J.-M.T.)
| |
Collapse
|
11
|
Wang C, Lau CY, Ma F, Zheng C. Genome-wide screen identifies curli amyloid fibril as a bacterial component promoting host neurodegeneration. Proc Natl Acad Sci U S A 2021; 118:e2106504118. [PMID: 34413194 PMCID: PMC8403922 DOI: 10.1073/pnas.2106504118] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Growing evidence indicates that gut microbiota play a critical role in regulating the progression of neurodegenerative diseases such as Parkinson's disease. The molecular mechanism underlying such microbe-host interaction is unclear. In this study, by feeding Caenorhabditis elegans expressing human α-syn with Escherichia coli knockout mutants, we conducted a genome-wide screen to identify bacterial genes that promote host neurodegeneration. The screen yielded 38 genes that fall into several genetic pathways including curli formation, lipopolysaccharide assembly, and adenosylcobalamin synthesis among others. We then focused on the curli amyloid fibril and found that genetically deleting or pharmacologically inhibiting the curli major subunit CsgA in E. coli reduced α-syn-induced neuronal death, restored mitochondrial health, and improved neuronal functions. CsgA secreted by the bacteria colocalized with α-syn inside neurons and promoted α-syn aggregation through cross-seeding. Similarly, curli also promoted neurodegeneration in C. elegans models of Alzheimer's disease, amyotrophic lateral sclerosis, and Huntington's disease and in human neuroblastoma cells.
Collapse
Affiliation(s)
- Chenyin Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chun Yin Lau
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Fuqiang Ma
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chaogu Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
12
|
A Gene Expression Signature to Predict Nucleotide Excision Repair Defects and Novel Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22095008. [PMID: 34066883 PMCID: PMC8125907 DOI: 10.3390/ijms22095008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
Nucleotide excision repair (NER) resolves DNA adducts, such as those caused by ultraviolet light. Deficient NER (dNER) results in a higher mutation rate that can predispose to cancer development and premature ageing phenotypes. Here, we used isogenic dNER model cell lines to establish a gene expression signature that can accurately predict functional NER capacity in both cell lines and patient samples. Critically, none of the identified NER deficient cell lines harbored mutations in any NER genes, suggesting that the prevalence of NER defects may currently be underestimated. Identification of compounds that induce the dNER gene expression signature led to the discovery that NER can be functionally impaired by GSK3 inhibition, leading to synergy when combined with cisplatin treatment. Furthermore, we predicted and validated multiple novel drugs that are synthetically lethal with NER defects using the dNER gene signature as a drug discovery platform. Taken together, our work provides a dynamic predictor of NER function that may be applied for therapeutic stratification as well as development of novel biological insights in human tumors.
Collapse
|
13
|
Niimi Y, Mizutani Y, Akiyama H, Watanabe H, Shiroki R, Hirabayashi Y, Hoshinaga K, Mutoh T. Cerebrospinal Fluid Profiles in Parkinson's Disease: No Accumulation of Glucosylceramide, but Significant Downregulation of Active Complement C5 Fragment. JOURNAL OF PARKINSONS DISEASE 2021; 11:221-232. [PMID: 33216044 DOI: 10.3233/jpd-202310] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND As mutations in glucocerebrosidase 1 (GBA1) are a major risk factor for Parkinson's disease (PD), decreased GBA1 activity might play an important role in the pathogenesis of the disease. However, there are currently no reports on glucosylceramide levels in the cerebrospinal fluid (CSF) in PD. OBJECTIVE We investigated whether glucosylceramide accumulation and abnormal immune status in the brain are associated with PD. METHODS We measured glucosylceramide by liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) as well as levels of the active fragment of complement C5, C5a, in the CSF of 33 PD, 15 amyotrophic lateral sclerosis (ALS) and 22 neurologically normal control (NNC) subjects. Serum C5a levels in all PD and ALS cases and in a limited number of NNC subjects (n = 8) were also measured. RESULTS C5a levels in CSF were significantly downregulated in PD compared with NNC. Moreover, CSF C5a/serum C5a ratio showed pronounced perturbations in PD and ALS patients. LC-ESI-MS/MS revealed a statistically significant accumulation of a specific subspecies of glucosylceramide (d18 : 1/C23 : 0 acyl chain fatty acid) in ALS, but not in PD. Interestingly, CSF glucosylceramide (d18 : 1/C23 : 0) exhibited a significant correlation with CSF C5a levels in PD, but not ALS. No correlation was observed between C5a levels or glucosylceramide subspecies content and disease duration, levodopa equivalent daily dose or Hoehn & Yahr staging in PD. CONCLUSION Our findings demonstrate complement dysregulation without glucosylceramide accumulation in PD CSF. Furthermore, we found an association between a specific glucosylceramide subspecies and immune status in PD.
Collapse
Affiliation(s)
- Yoshiki Niimi
- Department of Neurology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yasuaki Mizutani
- Department of Neurology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | | | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Ryoichi Shiroki
- Department of Urology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | | | - Kiyotaka Hoshinaga
- Department of Urology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Tatsuro Mutoh
- Department of Neurology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
14
|
Hu L, Dong MX, Huang YL, Lu CQ, Qian Q, Zhang CC, Xu XM, Liu Y, Chen GH, Wei YD. Integrated Metabolomics and Proteomics Analysis Reveals Plasma Lipid Metabolic Disturbance in Patients With Parkinson's Disease. Front Mol Neurosci 2020; 13:80. [PMID: 32714143 PMCID: PMC7344253 DOI: 10.3389/fnmol.2020.00080] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disease in the elderly with a pathogenesis that remains unclear. We aimed to explore its pathogenesis through plasma integrated metabolomics and proteomics analysis. The clinical data of consecutively recruited PD patients and healthy controls were assessed. Fasting plasma samples were obtained and analyzed using metabolomics and proteomics methods. After that, differentially expressed metabolites and proteins were identified for further bioinformatics analysis. No significant difference was found in the clinical data between these two groups. Eighty-three metabolites were differentially expressed in PD patients identified by metabolomics analysis. These metabolites were predominately lipid and lipid-like molecules (63%), among which 25% were sphingolipids. The sphingolipid metabolism pathway was enriched and tended to be activated in the following KEGG pathway analysis. According to the proteomics analysis, forty proteins were identified to be differentially expressed, seven of which were apolipoproteins. Furthermore, five of the six top ranking Gene Ontology terms from cellular components and eleven of the other fourteen Gene Ontology terms from biological processes were directly associated with lipid metabolism. In KEGG pathway analysis, the five enriched pathways were also significantly related with lipid metabolism (p < 0.05). Overall, Parkinson’s disease is associated with plasma lipid metabolic disturbance, including an activated sphingolipid metabolism and decreased apolipoproteins.
Collapse
Affiliation(s)
- Ling Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Neurology, Renmin Hospital of Wuhan University, Hubei General Hospital, Wuhan, China
| | - Mei-Xue Dong
- Department of Neurology, Renmin Hospital of Wuhan University, Hubei General Hospital, Wuhan, China
| | - Yan-Ling Huang
- Department of Neurology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China
| | - Chang-Qi Lu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Qian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-Cheng Zhang
- Department of Neurology, The People's Hospital of Tongliang District, Chongqing, China
| | - Xiao-Min Xu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guang-Hui Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Hubei General Hospital, Wuhan, China
| | - You-Dong Wei
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Bernal-Conde LD, Ramos-Acevedo R, Reyes-Hernández MA, Balbuena-Olvera AJ, Morales-Moreno ID, Argüero-Sánchez R, Schüle B, Guerra-Crespo M. Alpha-Synuclein Physiology and Pathology: A Perspective on Cellular Structures and Organelles. Front Neurosci 2020; 13:1399. [PMID: 32038126 PMCID: PMC6989544 DOI: 10.3389/fnins.2019.01399] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Alpha-synuclein (α-syn) is localized in cellular organelles of most neurons, but many of its physiological functions are only partially understood. α-syn accumulation is associated with Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy as well as other synucleinopathies; however, the exact pathomechanisms that underlie these neurodegenerative diseases remain elusive. In this review, we describe what is known about α-syn function and pathophysiological changes in different cellular structures and organelles, including what is known about its behavior as a prion-like protein. We summarize current knowledge of α-syn and its pathological forms, covering its effect on each organelle, including aggregation and toxicity in different model systems, with special interest on the mitochondria due to its relevance during the apoptotic process of dopaminergic neurons. Moreover, we explore the effect that α-syn exerts by interacting with chromatin remodeling proteins that add or remove histone marks, up-regulate its own expression, and resume the impairment that α-syn induces in vesicular traffic by interacting with the endoplasmic reticulum. We then recapitulate the events that lead to Golgi apparatus fragmentation, caused by the presence of α-syn. Finally, we report the recent findings about the accumulation of α-syn, indirectly produced by the endolysosomal system. In conclusion, many important steps into the understanding of α-syn have been made using in vivo and in vitro models; however, the time is right to start integrating observational studies with mechanistic models of α-syn interactions, in order to look at a more complete picture of the pathophysiological processes underlying α-synucleinopathies.
Collapse
Affiliation(s)
- Luis D. Bernal-Conde
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rodrigo Ramos-Acevedo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mario A. Reyes-Hernández
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Andrea J. Balbuena-Olvera
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ishbelt D. Morales-Moreno
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rubén Argüero-Sánchez
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Birgitt Schüle
- Department of Pathology, Stanford School of Medicine, Stanford University, Stanford, CA, United States
| | - Magdalena Guerra-Crespo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Medicina Regenerativa, Departamento de Cirugía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
16
|
Lukačišin M, Bollenbach T. Emergent Gene Expression Responses to Drug Combinations Predict Higher-Order Drug Interactions. Cell Syst 2019; 9:423-433.e3. [PMID: 31734160 DOI: 10.1016/j.cels.2019.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/03/2019] [Accepted: 10/11/2019] [Indexed: 01/10/2023]
Abstract
Effective design of combination therapies requires understanding the changes in cell physiology that result from drug interactions. Here, we show that the genome-wide transcriptional response to combinations of two drugs, measured at a rigorously controlled growth rate, can predict higher-order antagonism with a third drug in Saccharomyces cerevisiae. Using isogrowth profiling, over 90% of the variation in cellular response can be decomposed into three principal components (PCs) that have clear biological interpretations. We demonstrate that the third PC captures emergent transcriptional programs that are dependent on both drugs and can predict antagonism with a third drug targeting the emergent pathway. We further show that emergent gene expression patterns are most pronounced at a drug ratio where the drug interaction is strongest, providing a guideline for future measurements. Our results provide a readily applicable recipe for uncovering emergent responses in other systems and for higher-order drug combinations. A record of this paper's transparent peer review process is included in the Supplemental Information.
Collapse
Affiliation(s)
- Martin Lukačišin
- Institute for Biological Physics, University of Cologne, 50937 Cologne, Germany; IST Austria, 3400 Klosterneuburg, Austria
| | - Tobias Bollenbach
- Institute for Biological Physics, University of Cologne, 50937 Cologne, Germany.
| |
Collapse
|
17
|
Kakati T, Bhattacharyya DK, Barah P, Kalita JK. Comparison of Methods for Differential Co-expression Analysis for Disease Biomarker Prediction. Comput Biol Med 2019; 113:103380. [PMID: 31415946 DOI: 10.1016/j.compbiomed.2019.103380] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/01/2019] [Accepted: 08/03/2019] [Indexed: 01/23/2023]
Abstract
In the recent past, a number of methods have been developed for analysis of biological data. Among these methods, gene co-expression networks have the ability to mine functionally related genes with similar co-expression patterns, because of which such networks have been most widely used. However, gene co-expression networks cannot identify genes, which undergo condition specific changes in their relationships with other genes. In contrast, differential co-expression analysis enables finding co-expressed genes exhibiting significant changes across disease conditions. In this paper, we present some significant outcomes of a comparative study of four co-expression network module detection techniques, namely, THD-Module Extractor, DiffCoEx, MODA, and WGCNA, which can perform differential co-expression analysis on both gene and miRNA expression data (microarray and RNA-seq) and discuss the applications to Alzheimer's disease and Parkinson's disease research. Our observations reveal that compared to other methods, THD-Module Extractor is the most effective in finding modules with higher functional relevance and biological significance.
Collapse
Affiliation(s)
- Tulika Kakati
- Department of Computer Science and Engineering, Tezpur University, Tezpur, Assam, 784028, India
| | - Dhruba K Bhattacharyya
- Department of Computer Science and Engineering, Tezpur University, Tezpur, Assam, 784028, India.
| | - Pankaj Barah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Jugal K Kalita
- Department of Computer Science, University of Colorado, Colorado Springs, CO, 80918, USA
| |
Collapse
|
18
|
Olsen AL, Feany MB. Glial α-synuclein promotes neurodegeneration characterized by a distinct transcriptional program in vivo. Glia 2019; 67:1933-1957. [PMID: 31267577 DOI: 10.1002/glia.23671] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/29/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022]
Abstract
α-Synucleinopathies are neurodegenerative diseases that are characterized pathologically by α-synuclein inclusions in neurons and glia. The pathologic contribution of glial α-synuclein in these diseases is not well understood. Glial α-synuclein may be of particular importance in multiple system atrophy (MSA), which is defined pathologically by glial cytoplasmic α-synuclein inclusions. We have previously described Drosophila models of neuronal α-synucleinopathy, which recapitulate key features of the human disorders. We have now expanded our model to express human α-synuclein in glia. We demonstrate that expression of α-synuclein in glia alone results in α-synuclein aggregation, death of dopaminergic neurons, impaired locomotor function, and autonomic dysfunction. Furthermore, co-expression of α-synuclein in both neurons and glia worsens these phenotypes as compared to expression of α-synuclein in neurons alone. We identify unique transcriptomic signatures induced by glial as opposed to neuronal α-synuclein. These results suggest that glial α-synuclein may contribute to the burden of pathology in the α-synucleinopathies through a cell type-specific transcriptional program. This new Drosophila model system enables further mechanistic studies dissecting the contribution of glial and neuronal α-synuclein in vivo, potentially shedding light on mechanisms of disease that are especially relevant in MSA but also the α-synucleinopathies more broadly.
Collapse
Affiliation(s)
- Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Brennan S, Keon M, Liu B, Su Z, Saksena NK. Panoramic Visualization of Circulating MicroRNAs Across Neurodegenerative Diseases in Humans. Mol Neurobiol 2019; 56:7380-7407. [PMID: 31037649 PMCID: PMC6815273 DOI: 10.1007/s12035-019-1615-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and dementia pose one of the greatest health challenges this century. Although these NDs have been looked at as single entities, the underlying molecular mechanisms have never been collectively visualized to date. With the advent of high-throughput genomic and proteomic technologies, we now have the opportunity to visualize these diseases in a whole new perspective, which will provide a clear understanding of the primary and secondary events vital in achieving the final resolution of these diseases guiding us to new treatment strategies to possibly treat these diseases together. We created a knowledge base of all microRNAs known to be differentially expressed in various body fluids of ND patients. We then used several bioinformatic methods to understand the functional intersections and differences between AD, PD, ALS, and MS. These results provide a unique panoramic view of possible functional intersections between AD, PD, MS, and ALS at the level of microRNA and their cognate genes and pathways, along with the entities that unify and separate them. While the microRNA signatures were apparent for each ND, the unique observation in our study was that hsa-miR-30b-5p overlapped between all four NDS, and has significant functional roles described across NDs. Furthermore, our results also show the evidence of functional convergence of miRNAs which was associated with the regulation of their cognate genes represented in pathways that included fatty acid synthesis and metabolism, ECM receptor interactions, prion diseases, and several signaling pathways critical to neuron differentiation and survival, underpinning their relevance in NDs. Envisioning this group of NDs together has allowed us to propose new ways of utilizing circulating miRNAs as biomarkers and in visualizing diverse NDs more holistically . The critical molecular insights gained through the discovery of ND-associated miRNAs, overlapping miRNAs, and the functional convergence of microRNAs on vital pathways strongly implicated in neurodegenerative processes can prove immensely valuable in the identifying new generation of biomarkers, along with the development of miRNAs into therapeutics.
Collapse
Affiliation(s)
- Samuel Brennan
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Matthew Keon
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Bing Liu
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Zheng Su
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Nitin K. Saksena
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| |
Collapse
|
20
|
Patel D, Xu C, Nagarajan S, Liu Z, Hemphill WO, Shi R, Uversky VN, Caldwell GA, Caldwell KA, Witt SN. Alpha-synuclein inhibits Snx3-retromer-mediated retrograde recycling of iron transporters in S. cerevisiae and C. elegans models of Parkinson's disease. Hum Mol Genet 2019; 27:1514-1532. [PMID: 29452354 DOI: 10.1093/hmg/ddy059] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/12/2018] [Indexed: 01/31/2023] Open
Abstract
We probed the role of alpha-synuclein (α-syn) in modulating sorting nexin 3 (Snx3)-retromer-mediated recycling of iron transporters in Saccharomyces cerevisiae and Caenorhabditis elegans. In yeast, the membrane-bound heterodimer Fet3/Ftr1 is the high affinity iron importer. Fet3 is a membrane-bound multicopper ferroxidase, whose ferroxidase domain is orthologous to human ceruloplasmin (Cp), that oxidizes external Fe+2 to Fe+3; the Fe+3 ions then channel through the Ftr1 permease into the cell. When the concentration of external iron is low (<1 µM), Fet3/Ftr1 is maintained on the plasma membrane by retrograde endocytic-recycling; whereas, when the concentration of external iron is high (>10 µM), Fet3/Ftr1 is endocytosed and shunted to the vacuole for degradation. We discovered that α-syn expression phenocopies the high iron condition: under the low iron condition (<1 µM), α-syn inhibits Snx3-retromer-mediated recycling of Fet3/Ftr1 and instead shunts Fet3/Ftr1 into the multivesicular body pathway to the vacuole. α-Syn inhibits recycling by blocking the association of Snx3-mCherry molecules with endocytic vesicles, possibly by interfering with the binding of Snx3 to phosphatidylinositol-3-monophosphate. In C. elegans, transgenic worms expressing α-syn exhibit an age-dependent degeneration of dopaminergic neurons that is partially rescued by the iron chelator desferoxamine. This implies that α-syn-expressing dopaminergic neurons are susceptible to changes in iron neurotoxicity with age, whereby excess iron enhances α-syn-induced neurodegeneration. In vivo genetic analysis indicates that α-syn dysregulates iron homeostasis in worm dopaminergic neurons, possibly by inhibiting SNX-3-mediated recycling of a membrane-bound ortholog of Cp (F21D5.3), the iron exporter ferroportin (FPN1.1), or both.
Collapse
Affiliation(s)
- Dhaval Patel
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Chuan Xu
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Sureshbabu Nagarajan
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Zhengchang Liu
- Department of Biological Sciences, The University of New Orleans, New Orleans, LA 70148, USA
| | - Wayne O Hemphill
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Runhua Shi
- Department of Medicine, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Stephan N Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.,Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
21
|
Deryusheva E, Nemashkalova E, Galloux M, Richard C, Eléouët J, Kovacs D, Belle K, Tompa P, Uversky V, Permyakov S. Does Intrinsic Disorder in Proteins Favor Their Interaction with Lipids? Proteomics 2019; 19:e1800098. [DOI: 10.1002/pmic.201800098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 12/09/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Evgenia Deryusheva
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| | - Ekaterina Nemashkalova
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| | - Marie Galloux
- VIM, INRAUniversité Paris‐Saclay Jouy‐en‐Josas 78350 France
| | | | | | - Denis Kovacs
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
| | - Karo Belle
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
| | - Peter Tompa
- VIB, Structural Biology Research CenterVrije Universiteit Brussel Brussels 1050 Belgium
- Institute of EnzymologyResearch Centre for Natural Sciences of the Hungarian Academy of Sciences Budapest 1117 Hungary
| | - Vladimir Uversky
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research InstituteMorsani College of MedicineUniversity of South Florida Tampa FL 33612 USA
| | - Sergei Permyakov
- Russian Academy of SciencesInstitute for Biological Instrumentation Pushchino Moscow Region 142290 Russia
| |
Collapse
|
22
|
Pchelina S, Baydakova G, Nikolaev M, Senkevich K, Emelyanov A, Kopytova A, Miliukhina I, Yakimovskii A, Timofeeva A, Berkovich O, Fedotova E, Illarioshkin S, Zakharova E. Blood lysosphingolipids accumulation in patients with parkinson's disease with glucocerebrosidase 1 mutations. Mov Disord 2018; 33:1325-1330. [DOI: 10.1002/mds.27393] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 01/17/2023] Open
Affiliation(s)
- Sofya Pchelina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre (Kurchatov Institute); St. Petersburg Russian Federation
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
- Institute of Experimental Medicine; St. Petersburg Russian Federation
| | - Galina Baydakova
- Federal State Budgetary Institution (Research Centre for Medical Genetics); Moscow Russian Federation
| | - Mikhael Nikolaev
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre (Kurchatov Institute); St. Petersburg Russian Federation
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
| | - Konstantin Senkevich
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre (Kurchatov Institute); St. Petersburg Russian Federation
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
- Institute of Experimental Medicine; St. Petersburg Russian Federation
| | - Anton Emelyanov
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre (Kurchatov Institute); St. Petersburg Russian Federation
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
| | - Alena Kopytova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre (Kurchatov Institute); St. Petersburg Russian Federation
| | - Irina Miliukhina
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
- Institute of Experimental Medicine; St. Petersburg Russian Federation
| | - Andrey Yakimovskii
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
| | - Alla Timofeeva
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
| | - Olga Berkovich
- First Pavlov State Medical University of St. Petersburg; St. Petersburg Russian Federation
| | | | | | - Ekaterina Zakharova
- Federal State Budgetary Institution (Research Centre for Medical Genetics); Moscow Russian Federation
| |
Collapse
|
23
|
Ethanolamine and Phosphatidylethanolamine: Partners in Health and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4829180. [PMID: 28785375 PMCID: PMC5529665 DOI: 10.1155/2017/4829180] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022]
Abstract
Phosphatidylethanolamine (PE) is the second most abundant phospholipid in mammalian cells. PE comprises about 15–25% of the total lipid in mammalian cells; it is enriched in the inner leaflet of membranes, and it is especially abundant in the inner mitochondrial membrane. PE has quite remarkable activities: it is a lipid chaperone that assists in the folding of certain membrane proteins, it is required for the activity of several of the respiratory complexes, and it plays a key role in the initiation of autophagy. In this review, we focus on PE's roles in lipid-induced stress in the endoplasmic reticulum (ER), Parkinson's disease (PD), ferroptosis, and cancer.
Collapse
|
24
|
Aufschnaiter A, Kohler V, Diessl J, Peselj C, Carmona-Gutierrez D, Keller W, Büttner S. Mitochondrial lipids in neurodegeneration. Cell Tissue Res 2017; 367:125-140. [PMID: 27449929 PMCID: PMC5203858 DOI: 10.1007/s00441-016-2463-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/24/2016] [Indexed: 01/10/2023]
Abstract
Mitochondrial dysfunction is a common feature of many neurodegenerative diseases, including proteinopathies such as Alzheimer's or Parkinson's disease, which are characterized by the deposition of aggregated proteins in the form of insoluble fibrils or plaques. The distinct molecular processes that eventually result in mitochondrial dysfunction during neurodegeneration are well studied but still not fully understood. However, defects in mitochondrial fission and fusion, mitophagy, oxidative phosphorylation and mitochondrial bioenergetics have been linked to cellular demise. These processes are influenced by the lipid environment within mitochondrial membranes as, besides membrane structure and curvature, recruitment and activity of different proteins also largely depend on the respective lipid composition. Hence, the interaction of neurotoxic proteins with certain lipids and the modification of lipid composition in different cell compartments, in particular mitochondria, decisively impact cell death associated with neurodegeneration. Here, we discuss the relevance of mitochondrial lipids in the pathological alterations that result in neuronal demise, focussing on proteinopathies.
Collapse
Affiliation(s)
- Andreas Aufschnaiter
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
| | - Verena Kohler
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
| | - Jutta Diessl
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrheniusväg 20C, 106 91, Stockholm, Sweden
| | - Carlotta Peselj
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrheniusväg 20C, 106 91, Stockholm, Sweden
| | - Didac Carmona-Gutierrez
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
| | - Walter Keller
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010, Graz, Austria
| | - Sabrina Büttner
- Institute of Molecular Biosciences, University of Graz, Humboldtstraße 50, 8010, Graz, Austria.
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrheniusväg 20C, 106 91, Stockholm, Sweden.
| |
Collapse
|
25
|
Xing Y, Tang Y, Zhao L, Wang Q, Qin W, Ji X, Zhang J, Jia J. Associations between plasma ceramides and cognitive and neuropsychiatric manifestations in Parkinson's disease dementia. J Neurol Sci 2016; 370:82-87. [PMID: 27772793 DOI: 10.1016/j.jns.2016.09.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/16/2016] [Accepted: 09/16/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND The abnormal metabolism of ceramides may account for the pathogenesis of Parkinson's disease dementia (PDD). However, the effect of ceramides on cognitive domain impairments and neuropsychiatric symptoms of PDD remains unknown. METHODS A total of 38 PDD, 40 PD with no cognitive impairment (PD-NC) and 40 normal controls were included. A series of cognitive tests and the Neuropsychiatric Inventory (NPI) were used to assess cognitive domains and neuropsychiatric symptoms. A non-fasting blood sample was obtained from each subject. Plasma ceramide levels were tested by HPLC-MS/MS analysis. RESULTS C14:0 and C24:1 levels were significantly higher in PDD than in PD-NC and normal controls. Verbal memory was negatively correlated with C14:0 and C24:1. After controlling for confounding factors, C22:0, C20:0 and C18:0 were significantly associated with hallucinations, anxiety and sleep behavior disturbances, respectively. CONCLUSION In PDD, the increase in ceramide levels was correlated with decreased memory function and associated with higher odds of multiple neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Yi Xing
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Lina Zhao
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Qi Wang
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Wei Qin
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Ji
- Department of Cadre Health Care, Beijing Jishuitan Hospital, Beijing, China
| | - Jinlan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Jianping Jia
- Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, China; Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Sánchez Campos S, Rodríguez Diez G, Oresti GM, Salvador GA. Dopaminergic Neurons Respond to Iron-Induced Oxidative Stress by Modulating Lipid Acylation and Deacylation Cycles. PLoS One 2015; 10:e0130726. [PMID: 26076361 PMCID: PMC4468124 DOI: 10.1371/journal.pone.0130726] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
Metal-imbalance has been reported as a contributor factor for the degeneration of dopaminergic neurons in Parkinson Disease (PD). Specifically, iron (Fe)-overload and copper (Cu) mis-compartmentalization have been reported to be involved in the injury of dopaminergic neurons in this pathology. The aim of this work was to characterize the mechanisms of membrane repair by studying lipid acylation and deacylation reactions and their role in oxidative injury in N27 dopaminergic neurons exposed to Fe-overload and Cu-supplementation. N27 dopaminergic neurons incubated with Fe (1mM) for 24 hs displayed increased levels of reactive oxygen species (ROS), lipid peroxidation and elevated plasma membrane permeability. Cu-supplemented neurons (10, 50 μM) showed no evidence of oxidative stress markers. A different lipid acylation profile was observed in N27 neurons pre-labeled with [3H] arachidonic acid (AA) or [3H] oleic acid (OA). In Fe-exposed neurons, AA uptake was increased in triacylglycerols (TAG) whereas its incorporation into the phospholipid (PL) fraction was diminished. TAG content was 40% higher in Fe-exposed neurons than in controls. This increase was accompanied by the appearance of Nile red positive lipid bodies. Contrariwise, OA incorporation increased in the PL fractions and showed no changes in TAG. Lipid acylation profile in Cu-supplemented neurons showed AA accumulation into phosphatidylserine and no changes in TAG. The inhibition of deacylation/acylation reactions prompted an increase in oxidative stress markers and mitochondrial dysfunction in Fe-overloaded neurons. These findings provide evidence about the participation of lipid acylation mechanisms against Fe-induced oxidative injury and postulate that dopaminergic neurons cleverly preserve AA in TAG in response to oxidative stress.
Collapse
Affiliation(s)
- Sofía Sánchez Campos
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Guadalupe Rodríguez Diez
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Gerardo Martín Oresti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Gabriela Alejandra Salvador
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
- * E-mail:
| |
Collapse
|
27
|
Phosphatidylethanolamine deficiency disrupts α-synuclein homeostasis in yeast and worm models of Parkinson disease. Proc Natl Acad Sci U S A 2014; 111:E3976-85. [PMID: 25201965 DOI: 10.1073/pnas.1411694111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Phosphatidylserine decarboxylase, which is embedded in the inner mitochondrial membrane, synthesizes phosphatidylethanolamine (PE) and, in some cells, synthesizes the majority of this important phospholipid. Normal levels of PE can decline with age in the brain. Here we used yeast and worms to test the hypothesis that low levels of PE alter the homeostasis of the Parkinson disease-associated protein α-synuclein (α-syn). In yeast, low levels of PE in the phosphatidylserine decarboxylase deletion mutant (psd1Δ) cause decreased respiration, endoplasmic reticulum (ER) stress, a defect in the trafficking of the uracil permease, α-syn accumulation and foci, and a slow growth phenotype. Supplemental ethanolamine (ETA), which can be converted to PE via the Kennedy pathway enzymes in the ER, had no effect on respiration, whereas, in contrast, this metabolite partially eliminated ER stress, decreased α-syn foci formation, and restored growth close to that of wild-type cells. In Caenorhabditis elegans, RNAi depletion of phosphatidylserine decarboxylase in dopaminergic neurons expressing α-syn accelerates neurodegeneration, which supplemental ETA rescues. ETA fails to rescue this degeneration in worms that undergo double RNAi depletion of phosphatidylserine decarboxylase (psd-1) and choline/ETA phosphotransferase (cept-1), which encodes the last enzyme in the CDP-ETA Kennedy pathway. This finding suggests that ETA exerts its protective effect by boosting PE through the Kennedy pathway. Overall, a low level of PE causes ER stress, disrupts vesicle trafficking, and causes α-syn to accumulate; such cells likely die from a combination of ER stress and excessive accumulation of α-syn.
Collapse
|
28
|
da Silveira Dos Santos AX, Riezman I, Aguilera-Romero MA, David F, Piccolis M, Loewith R, Schaad O, Riezman H. Systematic lipidomic analysis of yeast protein kinase and phosphatase mutants reveals novel insights into regulation of lipid homeostasis. Mol Biol Cell 2014; 25:3234-46. [PMID: 25143408 PMCID: PMC4196872 DOI: 10.1091/mbc.e14-03-0851] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The regulatory pathways required to maintain eukaryotic lipid homeostasis are largely unknown. We developed a systematic approach to uncover new players in the regulation of lipid homeostasis. Through an unbiased mass spectrometry-based lipidomic screening, we quantified hundreds of lipid species, including glycerophospholipids, sphingolipids, and sterols, from a collection of 129 mutants in protein kinase and phosphatase genes of Saccharomyces cerevisiae. Our approach successfully identified known kinases involved in lipid homeostasis and uncovered new ones. By clustering analysis, we found connections between nutrient-sensing pathways and regulation of glycerophospholipids. Deletion of members of glucose- and nitrogen-sensing pathways showed reciprocal changes in glycerophospholipid acyl chain lengths. We also found several new candidates for the regulation of sphingolipid homeostasis, including a connection between inositol pyrophosphate metabolism and complex sphingolipid homeostasis through transcriptional regulation of AUR1 and SUR1. This robust, systematic lipidomic approach constitutes a rich, new source of biological information and can be used to identify novel gene associations and function.
Collapse
Affiliation(s)
- Aline Xavier da Silveira Dos Santos
- Department of Biochemistry, University of Geneva, Geneva CH-1211, Switzerland National Centre of Competence in Research "Chemical Biology,", University of Geneva, Geneva CH-1211, Switzerland
| | - Isabelle Riezman
- Department of Biochemistry, University of Geneva, Geneva CH-1211, Switzerland
| | - Maria-Auxiliadora Aguilera-Romero
- Department of Biochemistry, University of Geneva, Geneva CH-1211, Switzerland National Centre of Competence in Research "Chemical Biology,", University of Geneva, Geneva CH-1211, Switzerland
| | - Fabrice David
- École Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
| | - Manuele Piccolis
- Department of Molecular Biology, University of Geneva, Geneva CH-1211, Switzerland
| | - Robbie Loewith
- National Centre of Competence in Research "Chemical Biology,", University of Geneva, Geneva CH-1211, Switzerland Department of Molecular Biology, University of Geneva, Geneva CH-1211, Switzerland
| | - Olivier Schaad
- Department of Biochemistry, University of Geneva, Geneva CH-1211, Switzerland
| | - Howard Riezman
- Department of Biochemistry, University of Geneva, Geneva CH-1211, Switzerland National Centre of Competence in Research "Chemical Biology,", University of Geneva, Geneva CH-1211, Switzerland
| |
Collapse
|
29
|
Montefusco DJ, Matmati N, Hannun YA. The yeast sphingolipid signaling landscape. Chem Phys Lipids 2014; 177:26-40. [PMID: 24220500 PMCID: PMC4211598 DOI: 10.1016/j.chemphyslip.2013.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/18/2013] [Accepted: 10/19/2013] [Indexed: 12/13/2022]
Abstract
Sphingolipids are recognized as signaling mediators in a growing number of pathways, and represent potential targets to address many diseases. The study of sphingolipid signaling in yeast has created a number of breakthroughs in the field, and has the potential to lead future advances. The aim of this article is to provide an inclusive view of two major frontiers in yeast sphingolipid signaling. In the first section, several key studies in the field of sphingolipidomics are consolidated to create a yeast sphingolipidome that ranks nearly all known sphingolipid species by their level in a resting yeast cell. The second section presents an overview of most known phenotypes identified for sphingolipid gene mutants, presented with the intention of illuminating not yet discovered connections outside and inside of the field.
Collapse
Affiliation(s)
- David J Montefusco
- Dept. Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States.
| | - Nabil Matmati
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, United States
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, United States.
| |
Collapse
|
30
|
Haughey NJ, Zhu X, Bandaru VVR. A biological perspective of CSF lipids as surrogate markers for cognitive status in HIV. J Neuroimmune Pharmacol 2013; 8:1136-46. [PMID: 24203462 PMCID: PMC3909934 DOI: 10.1007/s11481-013-9506-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/03/2013] [Indexed: 12/14/2022]
Abstract
The development and application of biomarkers to neurodegenerative diseases has become increasingly important in clinical practice and therapeutic trials. While substantial progress has been made at the basic science level in understanding the pathophysiology of HIV-Associated Neurocognitive Disorders (HAND), there are significant limitations in our current ability to predict the onset or trajectory of disease, and to accurately determine the effects of therapeutic interventions. Thus, the development of objective biomarkers is critical to further our understanding and treatment of HAND. In recent years, biomarker discovery efforts have largely been driven forward through the implementation of multiple "omics" approaches that include (but are not restricted to): Lipidomics, proteomics, metabolomics, genomics, transcriptomics, and advances in brain imaging approaches such as functional connectomics. In this paper we summarize our progress to date on lipidomic approaches to biomarker discovery, discuss how these data have influenced basic research on the neuropathology of HAND, and implications for the development of therapeutics that target metabolic pathways involved in lipid handling.
Collapse
Affiliation(s)
- Norman J Haughey
- Department of Neurology, Division of Neuroimmunology and Neurological Infections, The Johns Hopkins University School of Medicine, Pathology 517, 600 North Wolfe Street, Baltimore, MD, 21287, USA,
| | | | | |
Collapse
|
31
|
Tardiff DF, Jui NT, Khurana V, Tambe MA, Thompson ML, Chung CY, Kamadurai HB, Kim HT, Lancaster AK, Caldwell KA, Caldwell GA, Rochet JC, Buchwald SL, Lindquist S. Yeast reveal a "druggable" Rsp5/Nedd4 network that ameliorates α-synuclein toxicity in neurons. Science 2013; 342:979-83. [PMID: 24158909 PMCID: PMC3993916 DOI: 10.1126/science.1245321] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
α-Synuclein (α-syn) is a small lipid-binding protein implicated in several neurodegenerative diseases, including Parkinson's disease, whose pathobiology is conserved from yeast to man. There are no therapies targeting these underlying cellular pathologies, or indeed those of any major neurodegenerative disease. Using unbiased phenotypic screens as an alternative to target-based approaches, we discovered an N-aryl benzimidazole (NAB) that strongly and selectively protected diverse cell types from α-syn toxicity. Three chemical genetic screens in wild-type yeast cells established that NAB promoted endosomal transport events dependent on the E3 ubiquitin ligase Rsp5/Nedd4. These same steps were perturbed by α-syn itself. Thus, NAB identifies a druggable node in the biology of α-syn that can correct multiple aspects of its underlying pathology, including dysfunctional endosomal and endoplasmic reticulum-to-Golgi vesicle trafficking.
Collapse
Affiliation(s)
- Daniel F Tardiff
- Whitehead Institute for Biomedical Research (WIBR), Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mielke MM, Maetzler W, Haughey NJ, Bandaru VVR, Savica R, Deuschle C, Gasser T, Hauser AK, Gräber-Sultan S, Schleicher E, Berg D, Liepelt-Scarfone I. Plasma ceramide and glucosylceramide metabolism is altered in sporadic Parkinson's disease and associated with cognitive impairment: a pilot study. PLoS One 2013; 8:e73094. [PMID: 24058461 PMCID: PMC3776817 DOI: 10.1371/journal.pone.0073094] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/17/2013] [Indexed: 01/08/2023] Open
Abstract
Background Mutations in the gene coding for glucocerebrosidase (GBA), which metabolizes glucosylceramide (a monohexosylceramide) into glucose and ceramide, is the most common genetic risk factor for sporadic Parkinson's disease (PD). GBA mutation carriers are more likely to have an earlier age of onset and to develop cognitive impairment and dementia. We hypothesized that plasma levels of lipids involved in ceramide metabolism would also be altered in PD non-GBA mutation carriers and associated with worse cognition. Methods Plasma ceramide, monohexosylceramide, and lactosylceramide levels in 26 cognitively normal PD patients, 26 PD patients with cognitive impairment or dementia, and 5 cognitively normal non-PD controls were determined by LC/ESI/MS/MS. Results Levels of all lipid species were higher in PD patients versus controls. Among PD patients, levels of ceramide C16:0, C18:0, C20:0, C22:0, and C24:1 and monohexosylceramide C16:0, C20:0 and C24:0 species were higher (all P<0.05) in those with versus without cognitive impairment. Conclusion These results suggest that plasma ceramide and monohexosylceramide metabolism is altered in PD non-GBA mutation carriers and that higher levels are associated with worse cognition. Additional studies with larger sample sizes, including cognitively normal controls, are needed to confirm these findings.
Collapse
Affiliation(s)
- Michelle M. Mielke
- Departments of Health Science Research and Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| | - Walter Maetzler
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Veera V. R. Bandaru
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Rodolfo Savica
- Departments of Health Science Research and Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Christian Deuschle
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Ann-Kathrin Hauser
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Susanne Gräber-Sultan
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Erwin Schleicher
- Department of Internal Medicine, University of Tuebingen, Tuebingen, Germany
| | - Daniela Berg
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| | - Inga Liepelt-Scarfone
- Hertie Institute for Clinical Brain Research, Center of Neurology, University of Tuebingen and DZNE, German Center for Neurodegenerative Diseases, Tuebingen, Germany
| |
Collapse
|
33
|
Zuo L, Motherwell MS. The impact of reactive oxygen species and genetic mitochondrial mutations in Parkinson's disease. Gene 2013; 532:18-23. [PMID: 23954870 DOI: 10.1016/j.gene.2013.07.085] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 07/23/2013] [Indexed: 12/27/2022]
Abstract
The exact pathogenesis of Parkinson's disease (PD) is still unknown and proper mechanisms that correspond to the disease remain unidentified. It is understood that PD is age-related; as age increases, the chance of onset responds accordingly. Although there are no current means of curing PD, the understanding of reactive oxygen species (ROS) provides significant insight to possible treatments. Complex I deficiencies of the respiratory chain account for the majority of unfavorable neural apoptosis generation in PD. Dopaminergic neurons are severely damaged as a result of the deficiency. Symptoms such as inhibited cognitive ability and loss of smooth motor function are the results of such impairment. The genetic mutations of Parkinson's related proteins such as PINK1 and LRRK2 contribute to mitochondrial dysfunction which precedes ROS formation. Various pathways are inhibited by these mutations, and inevitably causing neural cell damage. Antioxidants are known to negate the damaging effects of free radical overexpression. This paper expands on the specific impact of mitochondrial genetic change and production of free radicals as well as its correlation to the neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Li Zuo
- Molecular Physiology and Biophysics Laboratory, Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, Biophysics Graduate Program, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | | |
Collapse
|
34
|
Lee YJ, Shi R, Witt SN. The small molecule triclabendazole decreases the intracellular level of cyclic AMP and increases resistance to stress in Saccharomyces cerevisiae. PLoS One 2013; 8:e64337. [PMID: 23667708 PMCID: PMC3648474 DOI: 10.1371/journal.pone.0064337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 04/11/2013] [Indexed: 12/13/2022] Open
Abstract
The Ras-adenylyl cyclase-protein kinase A nutrient-sensing pathway controls metabolism, proliferation and resistance to stress in Saccharomyces cerevisiae. The genetic disruption of this pathway increases resistance to a variety of stresses. We show here that the pharmacological inhibition of this pathway by the drug triclabendazole increases resistance to oxidants, heat stress and extends the chronological life. Evidence is presented that triclabendazole decreases the intracellular level of cyclic AMP by inhibiting adenylyl cyclase and triggers the parallel rapid translocation of the stress-resistance transcription factor Msn2 from the cytosol into the nucleus, as deduced from experiments employing a strain in which MSN2 is replaced with MSN2-GFP (GFP, green fluorescent protein). Msn2 and Msn4 are responsible for activating the transcription of numerous genes that encode proteins that protect cells from stress. The results are consistent with triclabendazole either inhibiting the association of Ras with adenylyl cyclase or directly inhibiting adenylyl cyclase, which in turn triggers Msn2/4 to enter the nucleus and activate stress-responsible element gene expression.
Collapse
Affiliation(s)
- Yong Joo Lee
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana, United States of America
| | | | | |
Collapse
|
35
|
Matsumoto H, Shimizu Y, Tanaka A, Nogi T, Tabuchi I, Oyama K, Taniguchi M, Mannen H, Sasazaki S. The SNP in the promoter region of the bovine ELOVL5 gene influences economic traits including subcutaneous fat thickness. Mol Biol Rep 2012; 40:3231-7. [DOI: 10.1007/s11033-012-2398-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/17/2012] [Indexed: 12/31/2022]
|
36
|
α-Synuclein disrupts stress signaling by inhibiting polo-like kinase Cdc5/Plk2. Proc Natl Acad Sci U S A 2012; 109:16119-24. [PMID: 22988096 DOI: 10.1073/pnas.1206286109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Parkinson disease (PD) results from the slow, progressive loss of dopaminergic neurons in the substantia nigra. Alterations in α-synuclein (aSyn), such as mutations or multiplications of the gene, are thought to trigger this degeneration. Here, we show that aSyn disrupts mitogen-activated protein kinase (MAPK)-controlled stress signaling in yeast and human cells, which results in inefficient cell protective responses and cell death. aSyn is a substrate of the yeast (and human) polo-like kinase Cdc5 (Plk2), and elevated levels of aSyn prevent Cdc5 from maintaining a normal level of GTP-bound Rho1, which is an essential GTPase that regulates stress signaling. The nine N-terminal amino acids of aSyn are essential for the interaction with polo-like kinases. The results support a unique mechanism of PD pathology.
Collapse
|
37
|
Contribution of yeast models to neurodegeneration research. J Biomed Biotechnol 2012; 2012:941232. [PMID: 22910375 PMCID: PMC3403639 DOI: 10.1155/2012/941232] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/11/2012] [Accepted: 05/07/2012] [Indexed: 11/18/2022] Open
Abstract
As a model organism Saccharomyces cerevisiae has greatly contributed to our understanding of many fundamental aspects of cellular biology in higher eukaryotes. More recently, engineered yeast models developed to study endogenous or heterologous proteins that lay at the root of a given disease have become powerful tools for unraveling the molecular basis of complex human diseases like neurodegeneration. Additionally, with the possibility of performing target-directed large-scale screenings, yeast models have emerged as promising first-line approaches in the discovery process of novel therapeutic opportunities against these pathologies. In this paper, several yeast models that have contributed to the uncovering of the etiology and pathogenesis of several neurodegenerative diseases are described, including the most common forms of neurodegeneration worldwide, Alzheimer's, Parkinson's, and Huntington's diseases. Moreover, the potential input of these cell systems in the development of more effective therapies in neurodegeneration, through the identification of genetic and chemical suppressors, is also addressed.
Collapse
|
38
|
Oxidative stress in Alzheimer's and Parkinson's diseases: insights from the yeast Saccharomyces cerevisiae. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:132146. [PMID: 22701754 PMCID: PMC3371773 DOI: 10.1155/2012/132146] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 04/03/2012] [Accepted: 04/03/2012] [Indexed: 12/21/2022]
Abstract
Alzheimer's (AD) and Parkinson's (PD) diseases are the two most common causes of dementia in aged population. Both are protein-misfolding diseases characterized by the presence of protein deposits in the brain. Despite growing evidence suggesting that oxidative stress is critical to neuronal death, its precise role in disease etiology and progression has not yet been fully understood. Budding yeast Saccharomyces cerevisiae shares conserved biological processes with all eukaryotic cells, including neurons. This fact together with the possibility of simple and quick genetic manipulation highlights this organism as a valuable tool to unravel complex and fundamental mechanisms underlying neurodegeneration. In this paper, we summarize the latest knowledge on the role of oxidative stress in neurodegenerative disorders, with emphasis on AD and PD. Additionally, we provide an overview of the work undertaken to study AD and PD in yeast, focusing the use of this model to understand the effect of oxidative stress in both diseases.
Collapse
|
39
|
Triclabendazole protects yeast and mammalian cells from oxidative stress: identification of a potential neuroprotective compound. Biochem Biophys Res Commun 2011; 414:205-8. [PMID: 21946065 DOI: 10.1016/j.bbrc.2011.09.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 09/12/2011] [Indexed: 12/31/2022]
Abstract
The Prestwick and NIH chemical libraries were screened for drugs that protect baker's yeast from sugar-induced cell death (SICD). SICD is triggered when stationary-phase yeast cells are transferred from spent rich medium into water with 2% glucose and no other nutrients. The rapid, apoptotic cell death occurs because reactive oxygen species (ROS) accumulate. We found that triclabendazole, which is used to treat liver flukes in cattle and man, partially protects against SICD. Characterization of triclabendazole revealed that it also protects yeast cells from death induced by the Parkinson's disease-related protein alpha-synuclein (α-syn), which is known to induce the accumulation of ROS.
Collapse
|