1
|
Phongpreecha T, Mathi K, Cholerton B, Fox EJ, Sigal N, Espinosa C, Reincke M, Chung P, Hwang LJ, Gajera CR, Berson E, Perna A, Xie F, Shu CH, Hazra D, Channappa D, Dunn JE, Kipp LB, Poston KL, Montine KS, Maecker HT, Aghaeepour N, Montine TJ. Single-cell peripheral immunoprofiling of lewy body and Parkinson's disease in a multi-site cohort. Mol Neurodegener 2024; 19:59. [PMID: 39090623 PMCID: PMC11295553 DOI: 10.1186/s13024-024-00748-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Multiple lines of evidence support peripheral organs in the initiation or progression of Lewy body disease (LBD), a spectrum of neurodegenerative diagnoses that include Parkinson's Disease (PD) without or with dementia (PDD) and dementia with Lewy bodies (DLB). However, the potential contribution of the peripheral immune response to LBD remains unclear. This study aims to characterize peripheral immune responses unique to participants with LBD at single-cell resolution to highlight potential biomarkers and increase mechanistic understanding of LBD pathogenesis in humans. METHODS In a case-control study, peripheral mononuclear cell (PBMC) samples from research participants were randomly sampled from multiple sites across the United States. The diagnosis groups comprise healthy controls (HC, n = 159), LBD (n = 110), Alzheimer's disease dementia (ADD, n = 97), other neurodegenerative disease controls (NDC, n = 19), and immune disease controls (IDC, n = 14). PBMCs were activated with three stimulants (LPS, IL-6, and IFNa) or remained at basal state, stained by 13 surface markers and 7 intracellular signal markers, and analyzed by flow cytometry, which generated 1,184 immune features after gating. RESULTS The model classified LBD from HC with an AUROC of 0.87 ± 0.06 and AUPRC of 0.80 ± 0.06. Without retraining, the same model was able to distinguish LBD from ADD, NDC, and IDC. Model predictions were driven by pPLCγ2, p38, and pSTAT5 signals from specific cell populations under specific activation. The immune responses characteristic for LBD were not associated with other common medical conditions related to the risk of LBD or dementia, such as sleep disorders, hypertension, or diabetes. CONCLUSIONS AND RELEVANCE Quantification of PBMC immune response from multisite research participants yielded a unique pattern for LBD compared to HC, multiple related neurodegenerative diseases, and autoimmune diseases thereby highlighting potential biomarkers and mechanisms of disease.
Collapse
Affiliation(s)
- Thanaphong Phongpreecha
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Kavita Mathi
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | | | - Eddie J Fox
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Natalia Sigal
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Philip Chung
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Ling-Jen Hwang
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Eloise Berson
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Amalia Perna
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Feng Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Debapriya Hazra
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Divya Channappa
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jeffrey E Dunn
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Lucas B Kipp
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Fishman-Jacob T, Youdim MBH. A sporadic Parkinson's disease model via silencing of the ubiquitin-proteasome/E3 ligase component, SKP1A. J Neural Transm (Vienna) 2024; 131:675-707. [PMID: 37644186 PMCID: PMC11192832 DOI: 10.1007/s00702-023-02687-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Our and other's laboratory microarray-derived transcriptomic studies in human PD substantia nigra pars compacta (SNpc) samples have opened an avenue to concentrate on potential gene intersections or cross-talks along the dopaminergic (DAergic) neurodegenerative cascade in sporadic PD (SPD). One emerging gene candidate identified was SKP1A (p19, S-phase kinase-associated protein 1A), found significantly decreased in the SNpc as confirmed later at the protein level. SKP1 is part of the Skp1, Cullin 1, F-box protein (SCF) complex, the largest known class of sophisticated ubiquitin-proteasome/E3-ligases and was found to directly interact with FBXO7, a gene defective in PARK15-linked PD. This finding has led us to the hypothesis that a targeted site-specific reduction of Skp1 levels in DAergic neuronal cell culture and animal systems may result in a progressive loss of DAergic neurons and hopefully recreate motor disabilities in animals. The second premise considers the possibility that both intrinsic and extrinsic factors (e.g., manipulation of selected genes and mitochondria impairing toxins), alleged to play central roles in DAergic neurodegeneration in PD, may act in concert as modifiers of Skp1 deficiency-induced phenotype alterations ('dual-hit' hypothesis of neurodegeneration). To examine a possible role of Skp1 in DAergic phenotype, we have initially knocked down the expression of SKP1A gene in an embryonic mouse SN-derived cell line (SN4741) with short hairpin RNA (shRNA) lentiviruses (LVs). The deficiency of SKP1A closely recapitulated cardinal features of the DAergic pathology of human PD, such as decreased expression of DAergic phenotypic markers and cell cycle aberrations. Furthermore, the knocked down cells displayed a lethal phenotype when induced to differentiate exhibiting proteinaceous round inclusion structures, which were almost identical in composition to human Lewy bodies, a hallmark of PD. These findings support a role for Skp1 in neuronal phenotype, survival, and differentiation. The identification of Skp1 as a key player in DAergic neuron function suggested that a targeted site-specific reduction of Skp1 levels in mice SNpc may result in a progressive loss of DAergic neurons and terminal projections in the striatum. The injected LV SKP1shRNA to mouse SN resulted in decreased expression of Skp1 protein levels within DAergic neurons and loss of tyrosine hydroxylase immunoreactivity (TH-IR) in both SNpc and striatum that was accompanied by time-dependent motor disabilities. The reduction of the vertical movements, that is rearing, may be reminiscent of the early occurrence of hypokinesia and axial, postural instability in PD. According to the 'dual-hit' hypothesis of neurodegenerative diseases, it is predicted that gene-gene and/or gene-environmental factors would act in concert or sequentially to propagate the pathological process of PD. Our findings are compatible with this conjecture showing that the genetic vulnerability caused by knock down of SKP1A renders DAergic SN4741 cells especially sensitive to genetic reduction of Aldh1 and exposure to the external stressors MPP+ and DA, which have been implicated in PD pathology. Future consideration should be given in manipulation SKP1A expression as therapeutic window, via its induction genetically or pharmacological, to prevent degeneration of the nigra striatal dopamine neurons, since UPS is defective.
Collapse
Affiliation(s)
- Tali Fishman-Jacob
- Youdim Pharmaceutical Ltd, New Northern Industrial Park, 1 Ha- Tsmikha St, Stern Building, Fl-3, P. O. Box 72, 2069207, Yokneam, Israel
| | - Moussa B H Youdim
- Youdim Pharmaceutical Ltd, New Northern Industrial Park, 1 Ha- Tsmikha St, Stern Building, Fl-3, P. O. Box 72, 2069207, Yokneam, Israel.
| |
Collapse
|
3
|
Salas-Leal AC, Salas-Pacheco SM, Hernández-Cosaín EI, Vélez-Vélez LM, Antuna-Salcido EI, Castellanos-Juárez FX, Méndez-Hernández EM, Llave-León OL, Quiñones-Canales G, Arias-Carrión O, Sandoval-Carrillo AA, Salas-Pacheco JM. Differential expression of PSMC4, SKP1, and HSPA8 in Parkinson's disease: insights from a Mexican mestizo population. Front Mol Neurosci 2023; 16:1298560. [PMID: 38115821 PMCID: PMC10728481 DOI: 10.3389/fnmol.2023.1298560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative condition characterized by alpha-synuclein aggregation and dysfunctional protein degradation pathways. This study investigates the differential gene expression of pivotal components (UBE2K, PSMC4, SKP1, and HSPA8) within these pathways in a Mexican-Mestizo PD population compared to healthy controls. We enrolled 87 PD patients and 87 controls, assessing their gene expression levels via RT-qPCR. Our results reveal a significant downregulation of PSMC4, SKP1, and HSPA8 in the PD group (p = 0.033, p = 0.003, and p = 0.002, respectively). Logistic regression analyses establish a strong association between PD and reduced expression of PSMC4, SKP1, and HSPA8 (OR = 0.640, 95% CI = 0.415-0.987; OR = 0.000, 95% CI = 0.000-0.075; OR = 0.550, 95% CI = 0.368-0.823, respectively). Conversely, UBE2K exhibited no significant association or expression difference between the groups. Furthermore, we develop a gene expression model based on HSPA8, PSMC4, and SKP1, demonstrating robust discrimination between healthy controls and PD patients. Notably, the model's diagnostic efficacy is particularly pronounced in early-stage PD. In conclusion, our study provides compelling evidence linking decreased gene expression of PSMC4, SKP1, and HSPA8 to PD in the Mexican-Mestizo population. Additionally, our gene expression model exhibits promise as a diagnostic tool, particularly for early-stage PD diagnosis.
Collapse
Affiliation(s)
- Alma C. Salas-Leal
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Sergio M. Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Erik I. Hernández-Cosaín
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Lilia M. Vélez-Vélez
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | | | | | - Edna M. Méndez-Hernández
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - Osmel La Llave-León
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | | | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Ciudad de México, México
| | - Ada A. Sandoval-Carrillo
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| | - José M. Salas-Pacheco
- Instituto de Investigación Científica, Universidad Juárez del Estado de Durango, Durango, México
| |
Collapse
|
4
|
Liu J, Wang X, Ma R, Li T, Guo G, Ning B, Moran TH, Smith WW. AMPK signaling mediates synphilin-1-induced hyperphagia and obesity in Drosophila. J Cell Sci 2021; 134:jcs247742. [PMID: 33443093 PMCID: PMC7875497 DOI: 10.1242/jcs.247742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022] Open
Abstract
Expression of synphilin-1 in neurons induces hyperphagia and obesity in a Drosophila model. However, the molecular pathways underlying synphilin-1-linked obesity remain unclear. Here, Drosophila models and genetic tools were used to study the synphilin-1-linked pathways in energy balance by combining molecular biology and pharmacological approaches. We found that expression of human synphilin-1 in flies increased AMP-activated kinase (AMPK) phosphorylation at Thr172 compared with that in non-transgenic flies. Knockdown of AMPK reduced AMPK phosphorylation and food intake in non-transgenic flies, and further suppressed synphilin-1-induced AMPK phosphorylation, hyperphagia, fat storage and body weight gain in transgenic flies. Expression of constitutively activated AMPK significantly increased food intake and body weight gain in non-transgenic flies, but it did not alter food intake in the synphilin-1 transgenic flies. In contrast, expression of dominant-negative AMPK reduced food intake in both non-transgenic and synphilin-1 transgenic flies. Treatment with STO-609 also suppressed synphilin-1-induced AMPK phosphorylation, hyperphagia and body weight gain. These results demonstrate that the AMPK signaling pathway plays a critical role in synphilin-1-induced hyperphagia and obesity. These findings provide new insights into the mechanisms of synphilin-1-controlled energy homeostasis.
Collapse
Affiliation(s)
- Jingnan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Xiaobo Wang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rui Ma
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Tianxia Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Gongbo Guo
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Bo Ning
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Timothy H Moran
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanli W Smith
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Proteasome Subunits Involved in Neurodegenerative Diseases. Arch Med Res 2020; 52:1-14. [PMID: 32962866 DOI: 10.1016/j.arcmed.2020.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/25/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022]
Abstract
The ubiquitin-proteasome system is the major pathway for the maintenance of protein homeostasis. Its inhibition causes accumulation of ubiquitinated proteins; this accumulation has been associated with several of the most common neurodegenerative diseases. Several genetic factors have been identified for most neurodegenerative diseases, however, most cases are considered idiopathic, thus making the study of the mechanisms of protein accumulation a relevant field of research. It is often mentioned that the biggest risk factor for neurodegenerative diseases is aging, and several groups have reported an age-related alteration of the expression of some of the 26S proteasome subunits and a reduction of its activity. Proteasome subunits interact with proteins that are known to accumulate in neurodegenerative diseases such as α-synuclein in Parkinson's, tau in Alzheimer's, and huntingtin in Huntington's diseases. These interactions have been explored for several years, but only until recently, we are beginning to understand them. In this review, we discuss the known interactions, the underlying patterns, and the phenotypes associated with the 26S proteasome subunits in the etiology and progression of neurodegenerative diseases where there is evidence of proteasome involvement. Special emphasis is made in reviewing proteasome subunits that interact with proteins known to have an age-related altered expression or to be involved in neurodegenerative diseases to explore key effectors that may trigger or augment their progression. Interestingly, while the causes of age-related reduction of some of the proteasome subunits are not known, there are specific relationships between the observed neurodegenerative disease and the affected proteasome subunits.
Collapse
|
6
|
Synphilin-1 Interacts with AMPK and Increases AMPK Phosphorylation. Int J Mol Sci 2020; 21:ijms21124352. [PMID: 32570982 PMCID: PMC7352261 DOI: 10.3390/ijms21124352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/16/2023] Open
Abstract
A role for the cytoplasmic protein synphilin-1 in regulating energy balance has been demonstrated recently. Expression of synphilin-1 increases ATP levels in cultured cells. However, the mechanism by which synphilin-1 alters cellular energy status is unknown. Here, we used cell models and biochemical approaches to investigate the cellular functions of synphilin-1 on the AMP-activated protein kinase (AMPK) signaling pathway, which may affect energy balance. Overexpression of synphilin-1 increased AMPK phosphorylation (activation). Moreover, synphilin-1 interacted with AMPK by co-immunoprecipitation and GST (glutathione S-transferase) pull-down assays. Knockdown of synphilin-1 reduced AMPK phosphorylation. Overexpression of synphilin-1 also altered AMPK downstream signaling, i.e., a decrease in acetyl CoA carboxylase (ACC) phosphorylation, and an increase in p70S6K phosphorylation. Treatment of compound C (an AMPK inhibitor) reduced synphilin-1 binding with AMPK. In addition, compound C diminished synphilin-1-induced AMPK phosphorylation, and the increase in cellular ATP (adenosine triphosphate) levels. Our results demonstrated that synphilin-1 couples with AMPK, and they exert mutual effects on each other to regulate cellular energy status. These findings not only identify novel cellular actions of synphilin-1, but also provide new insights into the roles of synphilin-1 in regulating energy currency, ATP.
Collapse
|
7
|
Kitajima Y, Suzuki N, Nunomiya A, Osana S, Yoshioka K, Tashiro Y, Takahashi R, Ono Y, Aoki M, Nagatomi R. The Ubiquitin-Proteasome System Is Indispensable for the Maintenance of Muscle Stem Cells. Stem Cell Reports 2018; 11:1523-1538. [PMID: 30416048 PMCID: PMC6294073 DOI: 10.1016/j.stemcr.2018.10.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/06/2023] Open
Abstract
Adult muscle stem cells (satellite cells) are required for adult skeletal muscle regeneration. A proper balance between quiescence, proliferation, and differentiation is essential for the maintenance of the satellite cell pool and their regenerative function. Although the ubiquitin-proteasome is required for most protein degradation in mammalian cells, how its dysfunction affects tissue stem cells remains unclear. Here, we investigated the function of the proteasome in satellite cells using mice lacking the crucial proteasomal component, Rpt3. Ablation of Rpt3 in satellite cells decreased proteasome activity. Proteasome dysfunction in Rpt3-deficient satellite cells impaired their ability to proliferate, survive and differentiate, resulting in defective muscle regeneration. We found that inactivation of proteasomal activity induced proliferation defects and apoptosis in satellite cells. Mechanistically, insufficient proteasomal activity upregulated the p53 pathway, which caused cell-cycle arrest. Our findings delineate a critical function of the proteasome system in maintaining satellite cells in adult muscle. Ablation of Rpt3 in satellite cells leads to decreased proteasome activity Proteasome dysfunction in satellite cells results in defective muscle regeneration Proteasome dysfunction induces proliferation defects and apoptosis Inhibition of p53 rescues Rpt3-mediated defects in proliferation
Collapse
Affiliation(s)
- Yasuo Kitajima
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Basic and Translational Research Center for Hard Tissue Disease, 1-7-1 Sakamoto, Sakamoto, Nagasaki 852-8588, Japan; Japan Society for the Promotion of Science, Tokyo, Japan.
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Aki Nunomiya
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Shion Osana
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan
| | - Kiyoshi Yoshioka
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Basic and Translational Research Center for Hard Tissue Disease, 1-7-1 Sakamoto, Sakamoto, Nagasaki 852-8588, Japan
| | - Yoshitaka Tashiro
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Obu City, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yusuke Ono
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Basic and Translational Research Center for Hard Tissue Disease, 1-7-1 Sakamoto, Sakamoto, Nagasaki 852-8588, Japan.
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Ryoichi Nagatomi
- Division of Biomedical Engineering for Health and Welfare, Tohoku University Graduate School of Biomedical Engineering, 2-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
8
|
Differential Alterations in Metabolism and Proteolysis-Related Proteins in Human Parkinson's Disease Substantia Nigra. Neurotox Res 2017; 33:560-568. [PMID: 29218503 DOI: 10.1007/s12640-017-9843-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is the most common neurodegenerative disorder after Alzheimer's disease, with the majority of cases being sporadic or "idiopathic". The aetiology of the sporadic form is still unknown, but there is a broad consensus that Parkinson's disease involves multiple pathways. In previous human post-mortem studies investigating substantia nigra of parkinsonian subjects, gene expression alterations in various metabolic pathways including protein folding, trafficking, aggregation, ubiquitination and oxidative stress were found. These studies revealed transcriptomic dysregulation of various genes, amongst others Skp1A and PSMC4 (part of ubiquitin-proteasome system), HSC70 (belonging to the chaperone family) and ALDH1A1 (an enzyme involved in the catabolism of dopamine). To investigate whether these alterations are manifested at the protein level, we performed immunohistochemical analysis in the substantia nigra of Parkinson's disease and compared them to Alzheimer's disease and non-neurological post-mortem controls. We were able to confirm cell-specific reductions in the protein content of ALHD1A1 and Skp1A in the dopaminergic neurons of the substantia nigra of Parkinsonian patients compared to Alzheimer's and control subjects. Furthermore, we observed particular distribution for HSC70 and PSMC4 in the cytoplasm and accumulation within Lewy body in the dopaminergic neurons of the substantia nigra in Parkinson patients. These findings, together with previous evidence, suggest a malfunction of the ubiquitin-proteasome and possible autophagy systems as major players in protein misfolding and aggregation in Parkinson's disease. Nevertheless, this needs further proof, possibly with trajectory time line.
Collapse
|
9
|
Oh-Hashi K, Matsumoto S, Sakai T, Nomura Y, Okuda K, Nagasawa H, Hirata Y. Elucidating the rapid action of 2-(2-chlorophenyl)ethylbiguanide on HT-29 cells under a serum- and glucose-deprived condition. Cell Biol Toxicol 2017; 34:279-290. [PMID: 28871429 DOI: 10.1007/s10565-017-9410-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 08/21/2017] [Indexed: 10/18/2022]
Abstract
We recently demonstrated the cytotoxic action of a novel phenformin derivative, 2-(2-chlorophenyl)ethylbiguanide (2-Cl-Phen), on HT-29 cells under a serum- and glucose-deprived condition. In that study, we showed that the ATF6 arm of the ER stress pathway and c-Myc expression were downregulated 12 h after the treatment with 2-Cl-Phen. Through characterization of intracellular events at the early phase of the 2-Cl-Phen treatment before noticeable morphological changes, we found rapid fluctuations in the c-Myc and ATF4 proteins but not in their mRNAs in 2-Cl-Phen-treated HT-29 cells under the serum- and glucose-deprived condition. The 2-Cl-Phen-mediated downregulation of ATF4 protein was not paralleled by the phosphorylation status of PERK and eIF2α. Reduction of c-Myc expression by 2-Cl-Phen was more profound than that of ATF4 expression, and phosphorylated c-Myc was downregulated within 2 h. Pharmacological studies on the expression of c-Myc and ATF4 proteins showed that this decrease was mediated through proteasomal degradation but not by autophagy. Interestingly, treatment with lithium chloride, which is a well-known inhibitor of GSK3β, partially recovered the expression of ATF4 protein, but its effect on the level of total c-Myc protein was negligible. Treatment with 2-Cl-Phen increased the expression of phosphorylated AMPK, but Compound C, an AMPK inhibitor, did not influence the expression of c-Myc protein in HT-29 cells. Finally, we observed that 2-Cl-Phen partially attenuated the gene expression of integrin subunit α1 (ITGA1), a downstream target of c-Myc. Taken together, these results show that 2-Cl-Phen rapidly downregulated the expression of c-Myc in addition to ER stress responses in a post-translational manner. Further elucidation and improvement of this multi-target-directed compound will provide new insights for developing therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Kentaro Oh-Hashi
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan. .,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| | - Shiori Matsumoto
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Takayuki Sakai
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4, Daigakunishi, Gifu, 501-1196, Japan
| | - Yuki Nomura
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kensuke Okuda
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4, Daigakunishi, Gifu, 501-1196, Japan.,Laboratory of Bioorganic and Natural Products Chemistry, Kobe Pharmaceutical University, 4-19-1, Motoyama-kita, Higashinada, Kobe, 658-8558, Japan
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4, Daigakunishi, Gifu, 501-1196, Japan
| | - Yoko Hirata
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
10
|
Hypermethylation of Synphilin-1, Alpha-Synuclein-Interacting Protein (SNCAIP) Gene in the Cerebral Cortex of Patients with Sporadic Parkinson's Disease. Brain Sci 2017; 7:brainsci7070074. [PMID: 28653979 PMCID: PMC5532587 DOI: 10.3390/brainsci7070074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 11/17/2022] Open
Abstract
Objective: To determine and compare DNA methylation patterns between patients with Parkinson’s disease (PD) and age- and sex-similar matched non-PD controls. Background: Epigenetic regulation is one of the major mechanisms for an organism to respond to the environment through changes in gene expression and has been implicated in numerous disease processes. We would like to examine epigenetic modification patterns that may predispose or protect against PD. Methods: Frozen tissue samples of the human cerebral cortex from 12 PD patients and 12 subjects without PD pathology were obtained. Genome-wide DNA methylation profiling was performed using the Illumina HumanMethylation450 BeadChip array. Differential methylation was defined as a mean methylation level difference (delta β) of at least 0.20 (Δβ ≥ 0.20). Methylation regions with an absolute delta β value ≥ 0.20 were selected for further gene function studies. Results: We identified 2795 differentially methylated CpG sites in the frontal cortex of PD cases with a detection p-value of ≤ 0.01 and 328 differentially methylated CpG sites with a detection p-value of ≤ 0.001. A pattern of robust hypermethylation of synphilin-1, α-synuclein-interacting protein (SNCAIP) gene was found in the brain of PD cases (p = 4.93 × 10−7 and delta β = 0.60). Conclusion: Our findings support a link between SNCAIP methylation and PD risk. Hypomethylation of SNCAIP may function to protect against PD. The current results may suggest that the methylation status of SNCAIP could be useful as a marker in PD diagnosis and treatment and warrants further investigation.
Collapse
|
11
|
Translational and post-translational regulation of mouse cation transport regulator homolog 1. Sci Rep 2016; 6:28016. [PMID: 27302742 PMCID: PMC4908420 DOI: 10.1038/srep28016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 05/27/2016] [Indexed: 01/15/2023] Open
Abstract
Cation transport regulator homolog 1 (Chac1) is an endoplasmic reticulum (ER) stress inducible gene that has a function as a γ-glutamyl cyclotransferase involved in the degradation of glutathione. To characterize the translation and stability of Chac1, we found that the Kozak-like sequence present in the 5′ untranslated region (5′UTR) of the Chac1 mRNA was responsible for Chac1 translation. In addition, the short form (ΔChac1), which translated from the second ATG codon, was generated in the absence of the 5′UTR. The proteasome pathway predominantly participated in the stability of the Chac1 protein; however, its expression was remarkably up-regulated by co-transfection with ubiquitin genes. Using an immunoprecipitation assay, we revealed that ubiquitin molecule was directly conjugated to Chac1, and that mutated Chac1 with all lysine residues replaced by arginine was also ubiquitinated. Finally, we showed that WT Chac1 but not ΔChac1 reduced the intracellular level of glutathione. Taken together, our results suggest that the Chac1 protein expression is regulated in translational and post-translational fashion due to the Kozak-like sequence in the 5′UTR and the ubiquitin-mediated pathways. The bidirectional roles of ubiquitination in regulating Chac1 stabilization might give us a new insight into understanding the homeostasis of glutathione under pathophysiological conditions.
Collapse
|
12
|
Zaarur N, Xu X, Lestienne P, Meriin AB, McComb M, Costello CE, Newnam GP, Ganti R, Romanova NV, Shanmugasundaram M, Silva STN, Bandeiras TM, Matias PM, Lobachev KS, Lednev IK, Chernoff YO, Sherman MY. RuvbL1 and RuvbL2 enhance aggresome formation and disaggregate amyloid fibrils. EMBO J 2015; 34:2363-82. [PMID: 26303906 DOI: 10.15252/embj.201591245] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/13/2015] [Indexed: 02/02/2023] Open
Abstract
The aggresome is an organelle that recruits aggregated proteins for storage and degradation. We performed an siRNA screen for proteins involved in aggresome formation and identified novel mammalian AAA+ protein disaggregases RuvbL1 and RuvbL2. Depletion of RuvbL1 or RuvbL2 suppressed aggresome formation and caused buildup of multiple cytoplasmic aggregates. Similarly, downregulation of RuvbL orthologs in yeast suppressed the formation of an aggresome-like body and enhanced the aggregate toxicity. In contrast, their overproduction enhanced the resistance to proteotoxic stress independently of chaperone Hsp104. Mammalian RuvbL associated with the aggresome, and the aggresome substrate synphilin-1 interacted directly with the RuvbL1 barrel-like structure near the opening of the central channel. Importantly, polypeptides with unfolded structures and amyloid fibrils stimulated the ATPase activity of RuvbL. Finally, disassembly of protein aggregates was promoted by RuvbL. These data indicate that RuvbL complexes serve as chaperones in protein disaggregation.
Collapse
Affiliation(s)
- Nava Zaarur
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Xiaobin Xu
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA, USA
| | | | - Anatoli B Meriin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Mark McComb
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA, USA
| | - Catherine E Costello
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA, USA
| | - Gary P Newnam
- School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Rakhee Ganti
- School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nina V Romanova
- Laboratory of Amyloid Biology and Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Maruda Shanmugasundaram
- Department of Chemistry, University at Albany, State University of New York, Albany, NY, USA
| | - Sara T N Silva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | - Pedro M Matias
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Kirill S Lobachev
- School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Igor K Lednev
- Department of Chemistry, University at Albany, State University of New York, Albany, NY, USA
| | - Yury O Chernoff
- School of Biology, Georgia Institute of Technology, Atlanta, GA, USA Laboratory of Amyloid Biology and Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Michael Y Sherman
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Li X, Liu H, Fischhaber PL, Tang TS. Toward therapeutic targets for SCA3: Insight into the role of Machado-Joseph disease protein ataxin-3 in misfolded proteins clearance. Prog Neurobiol 2015; 132:34-58. [PMID: 26123252 DOI: 10.1016/j.pneurobio.2015.06.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/30/2015] [Accepted: 06/16/2015] [Indexed: 01/09/2023]
Abstract
Machado-Joseph disease (MJD, also known as spinocerebellar ataxia type 3, SCA3), an autosomal dominant neurological disorder, is caused by an abnormal expanded polyglutamine (polyQ) repeat in the ataxin-3 protein. The length of the expanded polyQ stretch correlates positively with the severity of the disease and inversely with the age at onset. To date, we cannot fully explain the mechanism underlying neurobiological abnormalities of this disease. Yet, accumulating reports have demonstrated the functions of ataxin-3 protein in the chaperone system, ubiquitin-proteasome system, and aggregation-autophagy, all of which suggest a role of ataxin-3 in the clearance of misfolded proteins. Notably, the SCA3 pathogenic form of ataxin-3 (ataxin-3(exp)) impairs the misfolded protein clearance via mechanisms that are either dependent or independent of its deubiquitinase (DUB) activity, resulting in the accumulation of misfolded proteins and the progressive loss of neurons in SCA3. Some drugs, which have been used as activators/inducers in the chaperone system, ubiquitin-proteasome system, and aggregation-autophagy, have been demonstrated to be efficacious in the relief of neurodegeneration diseases like Huntington's disease (HD), Parkinson's (PD), Alzheimer's (AD) as well as SCA3 in animal models and clinical trials, putting misfolded protein clearance on the list of potential therapeutic targets. Here, we undertake a comprehensive review of the progress in understanding the physiological functions of ataxin-3 in misfolded protein clearance and how the polyQ expansion impairs misfolded protein clearance. We then detail the preclinical studies targeting the elimination of misfolded proteins for SCA3 treatment. We close with future considerations for translating these pre-clinical results into therapies for SCA3 patients.
Collapse
Affiliation(s)
- Xiaoling Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongmei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Paula L Fischhaber
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, CA 91330-8262, USA.
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
14
|
Li T, Liu J, Smith WW. Synphilin-1 binds ATP and regulates intracellular energy status. PLoS One 2014; 9:e115233. [PMID: 25545246 PMCID: PMC4278857 DOI: 10.1371/journal.pone.0115233] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/19/2014] [Indexed: 11/19/2022] Open
Abstract
Recent studies have suggested that synphilin-1, a cytoplasmic protein, is involved in energy homeostasis. Overexpression of synphilin-1 in neurons results in hyperphagia and obesity in animal models. However, the mechanism by which synphilin-1 alters energy homeostasis is unknown. Here, we used cell models and biochemical approaches to investigate the cellular functions of synphilin-1 that may affect energy balance. Synphilin-1 was pulled down by ATP-agarose beads, and the addition of ATP and ADP reduced this binding, indicating that synphilin-1 bound ADP and ATP. Synphilin-1 also bound GMP, GDP, and GTP but with a lower affinity than it bound ATP. In contrast, synphilin-1 did not bind with CTP. Overexpression of synphilin-1 in HEK293T cells significantly increased cellular ATP levels. Genetic alteration to abolish predicted ATP binding motifs of synphilin-1 or knockdown of synphilin-1 by siRNA reduced cellular ATP levels. Together, these data demonstrate that synphilin-1 binds and regulates the cellular energy molecule, ATP. These findings provide a molecular basis for understanding the actions of synphilin-1 in energy homeostasis.
Collapse
Affiliation(s)
- Tianxia Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States of America
| | - Jingnan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States of America
| | - Wanli W. Smith
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States of America
- * E-mail:
| |
Collapse
|
15
|
Kitajima Y, Tashiro Y, Suzuki N, Warita H, Kato M, Tateyama M, Ando R, Izumi R, Yamazaki M, Abe M, Sakimura K, Ito H, Urushitani M, Nagatomi R, Takahashi R, Aoki M. Proteasome dysfunction induces muscle growth defects and protein aggregation. J Cell Sci 2014; 127:5204-17. [PMID: 25380823 PMCID: PMC4265737 DOI: 10.1242/jcs.150961] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The ubiquitin–proteasome and autophagy–lysosome pathways are the two major routes of protein and organelle clearance. The role of the proteasome pathway in mammalian muscle has not been examined in vivo. In this study, we report that the muscle-specific deletion of a crucial proteasomal gene, Rpt3 (also known as Psmc4), resulted in profound muscle growth defects and a decrease in force production in mice. Specifically, developing muscles in conditional Rpt3-knockout animals showed dysregulated proteasomal activity. The autophagy pathway was upregulated, but the process of autophagosome formation was impaired. A microscopic analysis revealed the accumulation of basophilic inclusions and disorganization of the sarcomeres in young adult mice. Our results suggest that appropriate proteasomal activity is important for muscle growth and for maintaining myofiber integrity in collaboration with autophagy pathways. The deletion of a component of the proteasome complex contributed to myofiber degeneration and weakness in muscle disorders that are characterized by the accumulation of abnormal inclusions.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yoshitaka Tashiro
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hitoshi Warita
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masaaki Kato
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Maki Tateyama
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Risa Ando
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Rumiko Izumi
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Maya Yamazaki
- Niigata University, Department of Cellular Neurobiology Brain Research Institute, Niigata 951-8510, Japan
| | - Manabu Abe
- Niigata University, Department of Cellular Neurobiology Brain Research Institute, Niigata 951-8510, Japan
| | - Kenji Sakimura
- Niigata University, Department of Cellular Neurobiology Brain Research Institute, Niigata 951-8510, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University Graduate School of Medicine, Wakayama 641-8510, Japan
| | - Makoto Urushitani
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Ryoichi Nagatomi
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
16
|
Hyttinen JM, Amadio M, Viiri J, Pascale A, Salminen A, Kaarniranta K. Clearance of misfolded and aggregated proteins by aggrephagy and implications for aggregation diseases. Ageing Res Rev 2014; 18:16-28. [PMID: 25062811 DOI: 10.1016/j.arr.2014.07.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/02/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Processing of misfolded proteins is important in order for the cell to maintain its normal functioning and homeostasis. Three systems control the quality of proteins: chaperone-mediated refolding, proteasomal degradation of ubiquitinated proteins, and finally, when the two others fail, aggrephagy, as selective form of autophagy, degrades ubiquitin-labelled aggregated cargos. In this route misfolded proteins gradually form larger aggregates, aggresomes and they eventually become double membrane-wrapped organelles called autophagosomes, which become degraded when they fuse to lysosomes, for reuse by the cell. The stages, the main molecules participating in the process, and the regulation of aggrephagy are discussed here, as is the role of protein aggregation in protein accumulation diseases. In particular, we emphasize that both Alzheimer's disease and age-related macular degeneration, two of the most common pathologies in the aged, are characterized by altered protein clearance and deposits. Based on the hypothesis that manipulations of autophagy may be potentially useful in these and other aggregation-related diseases, we will discuss some promising therapeutic strategies to counteract protein aggregates-induced cellular toxicity.
Collapse
|
17
|
Synphilin-1A is a phosphoprotein phosphatase 1-interacting protein and affects PPP1 sorting to subcellular compartments. J Mol Neurosci 2014; 55:385-95. [PMID: 24902662 DOI: 10.1007/s12031-014-0343-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/27/2014] [Indexed: 12/24/2022]
Abstract
Lewy bodies (LBs) are synphilin-1 (Sph1)-containing aggregates and histological hallmarks of Parkinson's disease. Therefore, understanding processes which modulate the aggregation of Sph1, or its isoform Sph1A, will contribute to our understanding of LBs formation. Protein phosphorylation promotes aggregation, but protein phosphatases with activity towards Sph1 have not been described. The present study documents the identification of a novel Sph1A/phosphoprotein phosphatase 1 (PPP1) complex and unravels its regulatory effect on Sph1A aggregation. Using yeast co-transformation and overlay blot assay, the interaction between Sph1A and PPP1 was mapped to the Sph1A RVTF motif. Then, Sph1A overexpression in human embryonic kidney 293 cells demonstrated that Sph1A specifically targets endogenous PPP1 isoforms to inclusion bodies and that Sph1A/PPP1 complex disruption enhances inclusion bodies formation. Finally, as Sph1A interacted with PPP1CC2, a PPP1 sperm-specific isoform, Sph1 and Sph1A expression was addressed in male germ cells by qRT-PCR, revealing high expression levels in round spermatids. Together, these observations established Sph1A as a novel PPP1-interacting protein able to affect PPP1 sorting to subcellular compartments and Sph1A/PPP1 complex as a negative modulator of LBs formation. Contrarily, in physiological conditions, Sph1 isoforms are pointed as putative participants in vesicle dynamics with implications in neurotransmission and spermiogenesis.
Collapse
|
18
|
Casadei N, Pöhler AM, Tomás-Zapico C, Torres-Peraza J, Schwedhelm I, Witz A, Zamolo I, De Heer R, Spruijt B, Noldus LPJJ, Klucken J, Lucas JJ, Kahle PJ, Krüger R, Riess O, Nuber S. Overexpression of synphilin-1 promotes clearance of soluble and misfolded alpha-synuclein without restoring the motor phenotype in aged A30P transgenic mice. Hum Mol Genet 2013; 23:767-81. [PMID: 24064336 DOI: 10.1093/hmg/ddt467] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lewy bodies and neurites are the pathological hallmark of Parkinson's disease. These structures are composed of fibrillized and ubiquitinated alpha-synuclein suggesting that impaired protein clearance is an important event in aggregate formation. The A30P mutation is known for its fast oligomerization, but slow fibrillization rate. Despite its toxicity to neurons, mechanisms involved in either clearance or conversion of A30P alpha-synuclein from its soluble state into insoluble fibrils and their effects in vivo are poorly understood. Synphilin-1 is present in Lewy bodies, interacting with alpha-synuclein in vivo and in vitro and promotes its sequestration into aggresomes, which are thought to act as cytoprotective agents facilitating protein degradation. We therefore crossed animals overexpressing A30P alpha-synuclein with synphilin-1 transgenic mice to analyze its impact on aggregation, protein clearance and phenotype progression. We observed that co-expression of synphilin-1 mildly delayed the motor phenotype caused by A30P alpha-synuclein. Additionally, the presence of N- and C-terminal truncated alpha-synuclein species and fibrils were strongly reduced in double-transgenic mice when compared with single-transgenic A30P mice. Insolubility of mutant A30P and formation of aggresomes was still detectable in aged double-transgenic mice, paralleled by an increase of ubiquitinated proteins and high autophagic activity. Hence, this study supports the notion that co-expression of synphilin-1 promotes formation of autophagic-susceptible aggresomes and consecutively the degradation of human A30P alpha-synuclein. Notably, although synphilin-1 overexpression significantly reduced formation of fibrils and astrogliosis in aged animals, a similar phenotype is present in single- and double-transgenic mice suggesting additional neurotoxic processes in disease progression.
Collapse
Affiliation(s)
- Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Tsai YC, Riess O, Soehn AS, Nguyen HP. The Guanine nucleotide exchange factor kalirin-7 is a novel synphilin-1 interacting protein and modifies synphilin-1 aggregate transport and formation. PLoS One 2012; 7:e51999. [PMID: 23284848 PMCID: PMC3527391 DOI: 10.1371/journal.pone.0051999] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/09/2012] [Indexed: 11/19/2022] Open
Abstract
Synphilin-1 has been identified as an interaction partner of α-synuclein, a key protein in the pathogenesis of Parkinson disease (PD). To further explore novel binding partners of synphilin-1, a yeast two hybrid screening was performed and kalirin-7 was identified as a novel interactor. We then investigated the effect of kalirin-7 on synphilin-1 aggregate formation. Coexpression of kalirin-7 and synphilin-1 caused a dramatic relocation of synphilin-1 cytoplasmic small inclusions to a single prominent, perinuclear inclusion. These perinuclear inclusions were characterized as being aggresomes according to their colocalization with microtubule organization center markers, and their formation was microtubule-dependent. Furthermore, kalirin-7 increased the susceptibility of synphilin-1 inclusions to be degraded as demonstrated by live cell imaging and quantification of aggregates. However, the kalirin-7-mediated synphilin-1 aggresome response was not dependent on the GEF activity of kalirin-7 since various dominant negative small GTPases could not inhibit the formation of aggresomes. Interestingly, the aggresome response was blocked by HDAC6 catalytic mutants and the HDAC inhibitor trichostatin A (TSA). Moreover, kalirin-7 decreased the level of acetylated α-tubulin in response to TSA, which suggests an effect of kalirin-7 on HDAC6-mediated protein transportation and aggresome formation. In summary, this is the first report demonstrating that kalirin-7 leads to the recruitment of synphilin-1 into aggresomes in a HDAC6-dependent manner and also links kalirin-7 to microtubule dynamics.
Collapse
Affiliation(s)
- Yu-Chun Tsai
- Department of Medical Genetics, University of Tuebingen, Tuebingen, Germany
| | - Olaf Riess
- Department of Medical Genetics, University of Tuebingen, Tuebingen, Germany
| | - Anne S. Soehn
- Department of Medical Genetics, University of Tuebingen, Tuebingen, Germany
| | - Huu Phuc Nguyen
- Department of Medical Genetics, University of Tuebingen, Tuebingen, Germany
- * E-mail:
| |
Collapse
|
20
|
Abstract
AIMS The pathogenesis of obesity remains incompletely understood. Drosophila have conserved neuroendocrine and digestion systems with human and become an excellent system for studying energy homeostasis. Here, we reported a novel obesity Drosophila model, in which expression of human protein, synphilin-1 (SP1), in neurons fosters positive energy balance. SUBJECTS AND METHODS To further understand the actions of SP1 in energy balance control, the upstream activation sequence UAS/GAL4 system was used to generate human SP1 transgenic Drosophila. We characterized a human SP1 transgenic Drosophila by assessing SP1 expression, fat lipid deposition, food intake and fly locomotor activity to determine the major behavioral changes and their consequences in the development of the obesity-like phenotype. RESULTS Overexpression of SP1 in neurons, but not peripheral cells, increased the body weight of flies compared with that of non-transgenic controls. SP1 increased food intake but did not affect locomotor activity. SP1 increased the levels of triacylglycerol, and the size of fat body cells and lipid droplets, indicating that SP1 increased lipid-fat disposition. Survival studies showed that SP1 transgenic flies were more resistant to food deprivation. SP1 regulated lipin gene expression that may participate in SP1-induced fat deposition and starvation resistance. CONCLUSION These studies demonstrate that SP1 expression affects energy homeostasis in ways that enhance positive energy balance and provide a useful obesity model for future pathogenesis and therapeutic studies.
Collapse
|
21
|
Lastres-Becker I, Ulusoy A, Innamorato NG, Sahin G, Rábano A, Kirik D, Cuadrado A. α-Synuclein expression and Nrf2 deficiency cooperate to aggravate protein aggregation, neuronal death and inflammation in early-stage Parkinson's disease. Hum Mol Genet 2012; 21:3173-92. [DOI: 10.1093/hmg/dds143] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
22
|
Mandel SA, Fishman-Jacob T, Youdim MB. Targeting Skp1, an Ubiquitin E3 Ligase Component Found Decreased in Sporadic Parkinsons Disease. NEURODEGENER DIS 2012; 10:220-3. [DOI: 10.1159/000333223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/12/2011] [Indexed: 11/19/2022] Open
|
23
|
Li X, Tamashiro KLK, Liu Z, Bello NT, Wang X, Aja S, Bi S, Ladenheim EE, Ross CA, Moran TH, Smith WW. A novel obesity model: synphilin-1-induced hyperphagia and obesity in mice. Int J Obes (Lond) 2011; 36:1215-21. [PMID: 22158267 PMCID: PMC3439552 DOI: 10.1038/ijo.2011.235] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aims The pathogenesis of obesity remains incompletely understood and the exploration of the role of novel proteins in obesity may provide important insights into its causes and treatments. Here we report a previously unidentified role for synphilin-1 in the controls of food intake and body weight. Synphilin-1, a cytoplasmic protein, was initially identified as an interaction partner of alpha-synuclein, and has implications in Parkinson's disease pathogenesis related to protein aggregation. Subjects and methods To study the in vivo role of synphilin-1, we characterized a human synphilin-1 transgenic mouse (SP1) by assessing synphilin-1 expression, plasma parameters, food intake and spontaneous activity to determine the major behavioral changes and their consequences in the development of the obesity phenotype. Results Expression of human synphilin-1 in brain neurons in SP1 mice resulted in increased food intake, body weight and body fat. SP1 mice also displayed hyperinsulinemia, hyperleptinemia and impaired glucose tolerance. Pair-feeding SP1 mice to amounts consumed by non-transgenic mice prevented the increased body weight, adiposity, hyperinsulinemia and hyperleptinemia demonstrating that these were all the consequences of increased food intake. Transgenic expression of synphilin-1 was enriched in hypothalamic nuclei involved in feeding control, and fasting induced elevated endogenous synphilin-1 levels at these sites, suggesting that synphilin-1 is an important player in the hypothalamic energy balance regulatory system. Conclusion These studies identify a novel function of synphilin-1 in controlling food intake and body weight, and may provide a unique obesity model for future studies of obesity pathogenesis and therapeutics.
Collapse
Affiliation(s)
- X Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Amoroso MR, Matassa DS, Laudiero G, Egorova AV, Polishchuk RS, Maddalena F, Piscazzi A, Paladino S, Sarnataro D, Garbi C, Landriscina M, Esposito F. TRAP1 and the proteasome regulatory particle TBP7/Rpt3 interact in the endoplasmic reticulum and control cellular ubiquitination of specific mitochondrial proteins. Cell Death Differ 2011; 19:592-604. [PMID: 21979464 DOI: 10.1038/cdd.2011.128] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Tumor necrosis factor receptor-associated protein-1 (TRAP1) is a mitochondrial (MITO) antiapoptotic heat-shock protein. The information available on the TRAP1 pathway describes just a few well-characterized functions of this protein in mitochondria. However, our group's use of mass-spectrometric analysis identified TBP7, an AAA-ATPase of the 19S proteasomal subunit, as a putative TRAP1-interacting protein. Surprisingly, TRAP1 and TBP7 colocalize in the endoplasmic reticulum (ER), as demonstrated by biochemical and confocal/electron microscopic analyses, and interact directly, as confirmed by fluorescence resonance energy transfer analysis. This is the first demonstration of TRAP1's presence in this cellular compartment. TRAP1 silencing by short-hairpin RNAs, in cells exposed to thapsigargin-induced ER stress, correlates with upregulation of BiP/Grp78, thus suggesting a role of TRAP1 in the refolding of damaged proteins and in ER stress protection. Consistently, TRAP1 and/or TBP7 interference enhanced stress-induced cell death and increased intracellular protein ubiquitination. These experiments led us to hypothesize an involvement of TRAP1 in protein quality control for mistargeted/misfolded mitochondria-destined proteins, through interaction with the regulatory proteasome protein TBP7. Remarkably, expression of specific MITO proteins decreased upon TRAP1 interference as a consequence of increased ubiquitination. The proposed TRAP1 network has an impact in vivo, as it is conserved in human colorectal cancers, is controlled by ER-localized TRAP1 interacting with TBP7 and provides a novel model of the ER-mitochondria crosstalk.
Collapse
Affiliation(s)
- M R Amoroso
- Department of Biochemistry and Medical Biotechnologies, University of Naples Federico II, Via Pansini 5, Naples 80131, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hernández-Vargas R, Fonseca-Ornelas L, López-González I, Riesgo-Escovar J, Zurita M, Reynaud E. Synphilin suppresses α-synuclein neurotoxicity in a Parkinson's disease Drosophila model. Genesis 2011; 49:392-402. [PMID: 21584925 DOI: 10.1002/dvg.20740] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder in humans. It affects 1% of the population over 65-years old. Its causes are environmental and genetic. As the world population ages, there is an urgent need for better and more detailed animal models for this kind of disease. In this work we show that the use of transgenic Drosophila is comparable to more complicated and costly animal models such as mice. The Drosophila model behaves very similar to the equivalent transgenic mice model. We show that both Synphilin-1 and α-synuclein are toxic by themselves, but when co-expressed, they suppress their toxicity reciprocally. Importantly, the symptoms induced in the fly can be treated and partially reverted using standard PD pharmacological treatments. This work showcases Drosophila as a detailed and multifaceted model for Parkinson's disease, providing a convenient platform in which to study and find new genetic modifiers of PD. genesis 49:392-402, 2011.
Collapse
Affiliation(s)
- René Hernández-Vargas
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | | | | | | | | |
Collapse
|
26
|
Alvarez-Castelao B, Castaño JG. Synphilin-1 inhibits alpha-synuclein degradation by the proteasome. Cell Mol Life Sci 2011; 68:2643-54. [PMID: 21103907 PMCID: PMC11114841 DOI: 10.1007/s00018-010-0592-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/13/2010] [Accepted: 11/04/2010] [Indexed: 01/01/2023]
Abstract
Intracellular deposits of aggregated alpha-synuclein are a hallmark of Parkinson's disease. Protein-protein interactions are critical in the regulation of cell proteostasis. Synphilin-1 interacts both in vitro and in vivo with alpha-synuclein promoting its aggregation. We report here that synphilin-1 specifically inhibits the degradation of alpha-synuclein wild-type and its missense mutants by the 20S proteasome due at least in part by the interaction of the ankyrin and coiled-coil domains of synphilin-1 (amino acids 331-555) with the N-terminal region (amino acids 1-60) of alpha-synuclein. Co-expression of synphilin-1 and alpha-synuclein wild-type in HeLa and N2A cells produces a specific increase in the half-life of alpha-synuclein, as degradation of unstable fluorescent reporters is not affected. Synphilin-1 inhibition can be relieved by co-expression of Siah-1 that targets synphilin-1 to degradation. Synphilin-1 inhibition of the proteasomal pathway of degradation of alpha-synuclein may help to understand the pathophysiological changes occurring in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Beatriz Alvarez-Castelao
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid y Consejo Superior de Investigaciones Científicas (UAM-CSIC), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) and Idipaz, Facultad de Medicina UAM, 28029 Madrid, Spain
| | - José G. Castaño
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid y Consejo Superior de Investigaciones Científicas (UAM-CSIC), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) and Idipaz, Facultad de Medicina UAM, 28029 Madrid, Spain
| |
Collapse
|
27
|
Büttner S, Delay C, Franssens V, Bammens T, Ruli D, Zaunschirm S, de Oliveira RM, Outeiro TF, Madeo F, Buée L, Galas MC, Winderickx J. Synphilin-1 enhances α-synuclein aggregation in yeast and contributes to cellular stress and cell death in a Sir2-dependent manner. PLoS One 2010; 5:e13700. [PMID: 21060871 PMCID: PMC2965147 DOI: 10.1371/journal.pone.0013700] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 10/07/2010] [Indexed: 11/19/2022] Open
Abstract
Background Parkinson's disease is characterized by the presence of cytoplasmic inclusions, known as Lewy bodies, containing both aggregated α-synuclein and its interaction partner, synphilin-1. While synphilin-1 is known to accelerate inclusion formation by α-synuclein in mammalian cells, its effect on cytotoxicity remains elusive. Methodology/Principal Findings We expressed wild-type synphilin-1 or its R621C mutant either alone or in combination with α-synuclein in the yeast Saccharomyces cerevisiae and monitored the intracellular localization and inclusion formation of the proteins as well as the repercussions on growth, oxidative stress and cell death. We found that wild-type and mutant synphilin-1 formed inclusions and accelerated inclusion formation by α-synuclein in yeast cells, the latter being correlated to enhanced phosphorylation of serine-129. Synphilin-1 inclusions co-localized with lipid droplets and endomembranes. Consistently, we found that wild-type and mutant synphilin-1 interacts with detergent-resistant membrane domains, known as lipid rafts. The expression of synphilin-1 did not incite a marked growth defect in exponential cultures, which is likely due to the formation of aggresomes and the retrograde transport of inclusions from the daughter cells back to the mother cells. However, when the cultures approached stationary phase and during subsequent ageing of the yeast cells, both wild-type and mutant synphilin-1 reduced survival and triggered apoptotic and necrotic cell death, albeit to a different extent. Most interestingly, synphilin-1 did not trigger cytotoxicity in ageing cells lacking the sirtuin Sir2. This indicates that the expression of synphilin-1 in wild-type cells causes the deregulation of Sir2-dependent processes, such as the maintenance of the autophagic flux in response to nutrient starvation. Conclusions/Significance Our findings demonstrate that wild-type and mutant synphilin-1 are lipid raft interacting proteins that form inclusions and accelerate inclusion formation of α-synuclein when expressed in yeast. Synphilin-1 thereby induces cytotoxicity, an effect most pronounced for the wild-type protein and mediated via Sir2-dependent processes.
Collapse
Affiliation(s)
- Sabrina Büttner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Charlotte Delay
- Functional Biology, Katholieke Universiteit Leuven, Heverlee, Belgium
- Alzheimer & Tauopathies, Inserm U837, Lille, France
- Jean-Pierre Aubert Research Centre, Université Lille Nord de France, Lille, France
- Faculté de Médecine-Pole Recherche, Université du Droit et de la Santé de Lille, Lille, France
- Centre Hospitalier Universitaire, Lille, France
| | - Vanessa Franssens
- Functional Biology, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Tine Bammens
- Functional Biology, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Doris Ruli
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Sandra Zaunschirm
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Rita Machado de Oliveira
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular, Lisboa, Portugal
- Instituto de Fisiologia, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago Fleming Outeiro
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular, Lisboa, Portugal
- Instituto de Fisiologia, Universidade de Lisboa, Lisboa, Portugal
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Luc Buée
- Alzheimer & Tauopathies, Inserm U837, Lille, France
- Jean-Pierre Aubert Research Centre, Université Lille Nord de France, Lille, France
- Faculté de Médecine-Pole Recherche, Université du Droit et de la Santé de Lille, Lille, France
- Centre Hospitalier Universitaire, Lille, France
| | - Marie-Christine Galas
- Alzheimer & Tauopathies, Inserm U837, Lille, France
- Jean-Pierre Aubert Research Centre, Université Lille Nord de France, Lille, France
- Faculté de Médecine-Pole Recherche, Université du Droit et de la Santé de Lille, Lille, France
- Centre Hospitalier Universitaire, Lille, France
| | - Joris Winderickx
- Functional Biology, Katholieke Universiteit Leuven, Heverlee, Belgium
- * E-mail:
| |
Collapse
|
28
|
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease that involves the deterioration of dopaminergic neurons in the substantia nigra pars compacta. Although the etiology of PD remains poorly understood, recent genetic, postmortem, and experimental evidence shows that abnormal protein accumulation and subsequent aggregate formation are prominent features of both sporadic and familial PD. While proteasome dysfunction is observed in PD, diverse mutations in the parkin gene are linked to early-onset autosomal-recessive forms of familial PD. We demonstrate that parkin, an E3 ubiquitin ligase, activates the 26S proteasome in an E3 ligase activity-independent manner. Furthermore, an N-terminal ubiquitin-like domain within parkin is critical for the activation of the 26S proteasome through enhancing the interaction between 19S proteasomal subunits, whereas the PD-linked R42P mutant abolishes this action. The current findings point to a novel role for parkin for 26S proteasome assembly and suggest that parkin mutations contribute to the proteasomal dysfunction in PD.
Collapse
|
29
|
Cuervo AM, Wong ESP, Martinez-Vicente M. Protein degradation, aggregation, and misfolding. Mov Disord 2010; 25 Suppl 1:S49-54. [PMID: 20187257 DOI: 10.1002/mds.22718] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The cellular surveillance systems guarantee proper removal of altered components from inside cells. Alterations of these systems in neurons have been proposed to be involved in the pathogenesis of different neurodegenerative disorders. In this review, we comment on the advances in our current understanding of how changes in the intracellular proteolytic systems, the main components of the cellular quality control system, contribute to neurodegeneration, with special emphasis on Parkinson's disease.
Collapse
Affiliation(s)
- Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | |
Collapse
|
30
|
Rogers N, Paine S, Bedford L, Layfield R. Review: the ubiquitin-proteasome system: contributions to cell death or survival in neurodegeneration. Neuropathol Appl Neurobiol 2010; 36:113-24. [PMID: 20202119 DOI: 10.1111/j.1365-2990.2010.01063.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The significance of the accumulation of ubiquitin-positive intraneuronal inclusions in the brains of those affected with different neurodegenerative diseases is currently unclear. While one interpretation is that the disease mechanism(s) involves dysfunction of an ubiquitin-mediated process, such as the ubiquitin-proteasome system, the inclusions are also found in surviving neurones, suggesting a possible neuroprotective role. Here we review recent evidence in support of these seemingly opposing notions gleaned from cell and animal models as well as investigations of patient samples, with particular emphasis on studies relevant to Parkinson's disease.
Collapse
Affiliation(s)
- N Rogers
- School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | | | | | | |
Collapse
|
31
|
Choi JS, Lee DH. CHIP promotes the degradation of mutant SOD1 by reducing its interaction with VCP and S6/S6′ subunits of 26S proteasome. Anim Cells Syst (Seoul) 2010. [DOI: 10.1080/19768351003765145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
32
|
Bukhatwa S, Zeng BY, Rose S, Jenner P. A comparison of changes in proteasomal subunit expression in the substantia nigra in Parkinson's disease, multiple system atrophy and progressive supranuclear palsy. Brain Res 2010; 1326:174-83. [PMID: 20176003 DOI: 10.1016/j.brainres.2010.02.045] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 02/10/2010] [Accepted: 02/11/2010] [Indexed: 11/29/2022]
Abstract
Dysfunction of the ubiquitin-proteasome system (UPS) occurs in dopaminergic neurones in the SN in PD and it is associated with Lewy body formation. However, it remains unknown whether this is specific to PD or whether it also occurs in multiple system atrophy (MSA) and progressive supranuclear palsy (PSP) where nigral dopaminergic neurones also degenerate. In the present study, we investigated changes in the expression of proteasomal subunits in the SN in PD, MSA and PSP. Immunohistochemistry double staining showed that proteasome 20S-alpha4 and -alpha6, and 20S-beta3 and -beta5i subunits are colocalized with tyrosine hydroxylase (TH)-positive cells in the SN of control, PD, MSA and PSP brain. Semi-quantitative analysis showed a significant loss of 20S-alpha4 and -alpha6 subunits TH-positive cells in PD, MSA and PSP compared to control tissue. There was no change in the expression of 20S-beta3 and -beta5i subunits in any of the disease states. The expression of PA700-Rpt5 subunits was not changed in PSP or PD but was significantly increased in MSA compared to control SN. PA700-Rpn10 subunit was not colocalized with TH within dopamine cells but was co-expressed with glial fibrillary acid protein (GFAP) positive astrocytes in the SN of all groups. PA28-alpha immunoreactivity was low in TH positive neurones in control tissue and quantification was not possible. Qualitative analysis suggested a decrease in PD and no immunoreactivity was detected in MSA or PSP. The results show that changes in proteasomal structure occur in the SN in PD, MSA and PSP and that these are similar in nature suggesting that dysfunction of UPS is not specific to PD or to Lewy body formation.
Collapse
Affiliation(s)
- Salma Bukhatwa
- School of Health and Biomedical Sciences, King's College, London, UK
| | | | | | | |
Collapse
|
33
|
Fishman-Jacob T, Reznichenko L, Youdim MBH, Mandel SA. A sporadic Parkinson disease model via silencing of the ubiquitin-proteasome/E3 ligase component SKP1A. J Biol Chem 2009; 284:32835-45. [PMID: 19748892 DOI: 10.1074/jbc.m109.034223] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to develop a new model of sporadic Parkinson disease (PD) based on silencing of the SKP1A gene, a component of the ubiquitin-proteasome/E3 ligase complex, Skp1, Cullin 1, F-box protein, which was found to be highly decreased in the substantia nigra of sporadic PD patients. Initially, an embryonic mouse substantia nigra-derived cell line (SN4741 cells) was infected with short hairpin RNA lentiviruses encoding the murine transcript of the SKP1A gene or with scrambled vector. SKP1A silencing resulted in increased susceptibility to neuronal damages induced by the parkinsonism-inducing neurotoxin 1-methyl-4-phenylpyridinium ion and serum starvation, in parallel with a decline in the expression of the dopaminergic markers, dopamine transporter and vesicular monoamine transporter-2. SKP1A-deficient cells presented a delay in completion of the cell cycle and the inability to arrest at the G(0)/G(1) phase when induced to differentiate. Instead, the cells progressed through S phase, developing rounded aggregates with characteristics of aggresomes including immunoreactivity for gamma-tubulin, alpha-synuclein, ubiquitin, tyrosine hydroxylase, Hsc-70 (70-kDa heat shock cognate protein), and proteasome subunit, and culminating in a lethal phenotype. Conversely, stably enforced expression of wild type SKP1A duplicated the survival index of naïve SN4741 cells under proteasomal inhibition injury, suggesting a new structural role of SKP1 in dopaminergic neuronal function, besides its E3 ligase activity. These results link, for the first time, SKP1 to dopamine neuronal function and survival, suggesting an essential role in sporadic PD. In summary, this new model has reproduced to a significant extent the molecular alterations described in sporadic PD at the cellular level, implicating Skp1 as a potential modifier in sporadic PD neurodegeneration.
Collapse
Affiliation(s)
- Tali Fishman-Jacob
- Eve Topf and National Parkinson Foundation Centers for Neurodegenerative Diseases and the Department of Molecular Pharmacology, Faculty of Medicine, Technion, 31096 Haifa, Israel
| | | | | | | |
Collapse
|
34
|
Cook C, Petrucelli L. A critical evaluation of the ubiquitin-proteasome system in Parkinson's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1792:664-75. [PMID: 19419700 PMCID: PMC2828612 DOI: 10.1016/j.bbadis.2009.01.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 01/12/2009] [Accepted: 01/27/2009] [Indexed: 01/04/2023]
Abstract
The evidence for impairment in the ubiquitin proteasome system (UPS) in Parkinson's disease (PD) is mounting and becoming increasingly more convincing. However, it is presently unclear whether UPS dysfunction is a cause or result of PD pathology, a crucial distinction which impedes both the understanding of disease pathogenesis and the development of effectual therapeutic approaches. Recent findings discussed within this review offer new insight and provide direction for future research to conclusively resolve this debate.
Collapse
Affiliation(s)
- Casey Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | |
Collapse
|
35
|
Diaz-Corrales FJ, Asanuma M, Miyazaki I, Miyoshi K, Hattori N, Ogawa N. Dopamine induces supernumerary centrosomes and subsequent cell death through Cdk2 up-regulation in dopaminergic neuronal cells. Neurotox Res 2009; 14:295-305. [PMID: 19073433 DOI: 10.1007/bf03033854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aggregation of proteins in the centrosome is implicated in the pathophysiology of Parkinson's disease. However, the relevance of the centrosome in neurodegeneration is still obscure. Centrosome duplication is initiated by the cyclin E/cyclin-dependent kinase 2 (Cdk2) complex. The present study determined changes in cyclin E or Cdk2 expression and in the centrosomal structure in dopaminergic neuronal CATH.a cells exposed to 50, 100 and 150 micromolar dopamine (DA) for 24 h. DA induced significant increase in Cdk2 protein and cyclin E protein, but not cyclin e mRNA. In DA-treated cells, the intense cyclin E- and Cdk2-immunofluorescence signals were co-localized around large and supernumerary centrosomes, and these two parameters of centrosome amplification were significantly increased compared with the control. Simultaneous co-treatment with DA and a Cdk2 inhibitor blocked centrosome amplification and enhanced cell viability. Our results demonstrated that DA could lead to cyclin E accumulation and Cdk2 up-regulation triggering supernumerary centrosomes and apoptotic cell death.
Collapse
Affiliation(s)
- Francisco J Diaz-Corrales
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Molecular pathology of Lewy body diseases. Int J Mol Sci 2009; 10:724-45. [PMID: 19399218 PMCID: PMC2671999 DOI: 10.3390/ijms10030724] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 02/03/2009] [Accepted: 02/23/2009] [Indexed: 11/16/2022] Open
Abstract
Lewy body diseases are characterized by the presence of Lewy bodies, alpha-synuclein(AS)-positive inclusions in the brain. Since their main component is conformationally modified AS, aggregation of the latter is thought to be a key pathogenic event in these diseases. The analysis of inclusion body constituents gives additional information about pathways also involved in the pathology of synucleinopathies. Widespread mitochondrial dysfunction is very closely related to disease development. The impairment of protein degradation pathways, including both the ubiquitin-proteasome system and the autophagy-lysosome pathway also play an important role during the development of Lewy body diseases. Finally, differential expression changes of isoforms corresponding to genes primarily involved in Lewy body formation point to alternative splicing as another important mechanism in the development of Parkinson’s disease, as well as dementia with Lewy bodies. The present paper attempts to give an overview of recent molecular findings related to the pathogenesis of Lewy body diseases.
Collapse
|
37
|
Jin HG, Yamashita H, Nakamura T, Fukuba H, Takahashi T, Hiji M, Kohriyama T, Matsumoto M. Synphilin-1 transgenic mice exhibit mild motor impairments. Neurosci Lett 2008; 445:12-7. [PMID: 18782602 DOI: 10.1016/j.neulet.2008.08.073] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Revised: 08/24/2008] [Accepted: 08/27/2008] [Indexed: 11/28/2022]
Abstract
Synphilin-1 represents a cytoplasmic protein that interacts with alpha-synuclein and localizes close to synaptic vesicles. The interaction of synphilin-1 with several proteins involved in Parkinson's disease suggests that it might be involved in the pathogenesis of the disease. Nonetheless, the function of synphilin-1 remains unclear. In the present study, we generated transgenic mice expressing human synphilin-1 under the prion protein promoter. Synphilin-1 was widely expressed in neurons in the brain including the substantia nigra, where massive loss of dopamine neurons was not observed. In the transgenic mouse brain, synphilin-1 protein was polyubiquitinated, and partially insoluble. Although modified-SHIRPA revealed no significant difference in behavior and morphology, the reduced rotarod performance and step length were observed in transgenic mice as compared with non-transgenic littermates. Synphilin-1 might be involved in motor function, and its accumulation in the central nervous system can cause motor impairments.
Collapse
Affiliation(s)
- Hong-Guo Jin
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical Sciences, 1-2-3 Kasumi, Hiroshima 734-8551, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Zaarur N, Meriin AB, Gabai VL, Sherman MY. Triggering aggresome formation. Dissecting aggresome-targeting and aggregation signals in synphilin 1. J Biol Chem 2008; 283:27575-27584. [PMID: 18635553 DOI: 10.1074/jbc.m802216200] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abnormal polypeptides that escape proteasome-dependent degradation and aggregate in cytosol can be transported via microtubules to an aggresome, a recently discovered organelle where aggregated proteins are stored or degraded by autophagy. We used synphilin 1, a protein implicated in Parkinson disease, as a model to study mechanisms of aggresome formation. When expressed in naïve HEK293 cells, synphilin 1 forms multiple small highly mobile aggregates. However, proteasome or Hsp90 inhibition rapidly triggered their translocation into the aggresome, and surprisingly, this response was independent on the expression level of synphilin 1. Therefore, aggresome formation, but not aggregation of synphilin 1, represents a special cellular response to a failure of the proteasome/chaperone machinery. Importantly, translocation to aggresomes required a special aggresome-targeting signal within the sequence of synphilin 1, an ankyrin-like repeat domain. On the other hand, formation of multiple small aggregates required an entirely different segment within synphilin 1, indicating that aggregation and aggresome formation determinants can be separated genetically. Furthermore, substitution of the ankyrin-like repeat in synphilin 1 with an aggresome-targeting signal from huntingtin was sufficient for aggresome formation upon inhibition of the proteasome. Analogously, attachment of the ankyrin-like repeat to a huntingtin fragment lacking its aggresome-targeting signal promoted its transport to aggresomes. These findings indicate the existence of transferable signals that target aggregation-prone polypeptides to aggresomes.
Collapse
Affiliation(s)
- Nava Zaarur
- Department of Biochemistry, Boston University Medical School, Boston, Massachusetts 02118
| | - Anatoli B Meriin
- Department of Biochemistry, Boston University Medical School, Boston, Massachusetts 02118
| | - Vladimir L Gabai
- Department of Biochemistry, Boston University Medical School, Boston, Massachusetts 02118
| | - Michael Y Sherman
- Department of Biochemistry, Boston University Medical School, Boston, Massachusetts 02118.
| |
Collapse
|
39
|
Wahl C, Kautzmann S, Krebiehl G, Strauss K, Woitalla D, Müller T, Bauer P, Riess O, Krüger R. A comprehensive genetic study of the proteasomal subunit S6 ATPase in German Parkinson's disease patients. J Neural Transm (Vienna) 2008; 115:1141-8. [PMID: 18446261 DOI: 10.1007/s00702-008-0054-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 04/10/2008] [Indexed: 12/12/2022]
Abstract
Dysfunction of proteasomal protein degradation is involved in neurodegeneration in Parkinson's disease (PD). Recently we identified the regulatory proteasomal subunit S6 ATPase as a novel interactor of synphilin-1, which is a substrate of the ubiquitin-ligase Parkin (PARK2) and an interacting protein of alpha-synuclein (PARK1). To further investigate a potential role in the pathogenesis of PD, we performed a detailed mutation analysis of the S6 ATPase gene in a large sample of 486 German sporadic and familial PD patients. Direct sequencing revealed two novel intronic variants. An insertion/deletion variant in intron 5 of the S6 ATPase gene was more frequent in patients compared to controls. Moreover, this variant was significantly more frequent in early-onset compared to late-onset PD patients. The identification of a genetic link between a regulatory proteasomal subunit and PD further underscores the relevance of disturbed protein degradation in PD.
Collapse
Affiliation(s)
- Claudia Wahl
- Laboratory of Functional Neurogenomics, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tubingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Di Napoli M, Shah IM, Stewart DA. Molecular pathways and genetic aspects of Parkinson's disease: from bench to bedside. Expert Rev Neurother 2008; 7:1693-729. [PMID: 18052765 DOI: 10.1586/14737175.7.12.1693] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Idiopathic Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by dopaminergic neuronal loss within the substantia nigra. The incidence and prevalence of PD is rising with an increasing aging population. PD is a slowly progressive condition and patients can develop debilitating motor and functional impairment. Current research has implicated oxidative stress, alpha-synucleinopathy and dysfunction of the ubiquitin-proteasome system in the pathogenesis of PD. A number of gene mutations have also been linked to the development of PD. The elucidation of these new molecular pathways has increased our knowledge of PD pathophysiology. This article reviews important molecular mechanisms and genetic causes implicated in the pathogenesis of PD, which has led to new areas of therapeutic drug research.
Collapse
Affiliation(s)
- Mario Di Napoli
- Neurological Service, San Camillo de'Lellis General Hospital, I-2100 Rieti, Italy.
| | | | | |
Collapse
|
41
|
Abstract
Accumulation of misfolded proteins in proteinaceous inclusions is a prominent pathological feature common to many age-related neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. In cultured cells, when the production of misfolded proteins exceeds the capacity of the chaperone refolding system and the ubiquitin-proteasome degradation pathway, misfolded proteins are actively transported to a cytoplasmic juxtanuclear structure called an aggresome. Aggresome formation is recognized as a cytoprotective response serving to sequester potentially toxic misfolded proteins and facilitate their clearance by autophagy. Recent evidence indicates that aggresome formation is mediated by dynein/dynactin-mediated microtubule-based transport of misfolded proteins to the centrosome and involves several regulators, including histone deacetylase 6, E3 ubiquitin-protein ligase parkin, deubiquitinating enzyme ataxin-3, and ubiquilin-1. Characterization of the molecular mechanisms underlying aggresome formation and its regulation has begun to provide promising therapeutic targets that may be relevant to neurodegenerative diseases. In this review, we provide an overview of the molecular machinery controlling aggresome formation and discuss potential useful compounds and intervention strategies for preventing or reducing the cytotoxicity of misfolded and aggregated proteins.
Collapse
Affiliation(s)
- J.A. Olzmann
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - L. Li
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - L.S. Chin
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
42
|
Abstract
The past 10 years has seen a shift in our etiological concepts of Parkinson's disease, moving from a nearly exclusively environmentally mediated disease towards a complex disorder with important genetic contributors. The identification of responsible mutations in certain genes, particularly alpha-synuclein, Parkin, PINK1, DJ-1 and LRRK2, has increased our understanding of the clinical and pathological changes underlying Parkinson's disease, with implications for patient diagnosis, management and future research. This review will outline the specific genetic advances, discuss their implications for clinical practice and hint at future directions for research into this common and disabling disease.
Collapse
Affiliation(s)
- Michael R Douglas
- University of Birmingham Medical School, Vincent Drive, Edgbaston, Birmingham, B15 2TT, UK.
| | | | | |
Collapse
|