1
|
Huang KY, Yu HC, Lu MC, Tseng HYH, Shen JJ, Lin CY, Chen PC, Shen YT, Chung PR, Tsai HK, Zhou SR, Wang CL, Lai NS, Lin TH, Huang HB. Identification of a novel Eps 15 homology domain-containing protein 1 (EHD1) and EHD4-binding motif in phostensin. J Biochem 2025; 177:297-304. [PMID: 39776131 DOI: 10.1093/jb/mvaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Phostensin (PTS) encoded by KIAA1949 binds to protein phosphatase 1, F-actin, Eps 15 homology domain-containing protein 1 (EHD1) and EHD4. Most EHD-binding proteins contain a consensus motif, Asn-Pro-Phe (NPF), which interacts with the C-terminal EH domain of EHD proteins. Nevertheless, the NPF motif is absent in PTS. The binding motif for PTS to interact with EHD1 (or EHD4) remains unknown. Here, we identified that PTS-α binds to EHD1 (or EHD4) through the region of residues 51-80, which contains a consensus motif, 64ILV(X)4(L/V)RL74S. This novel consensus motif is also found in vacuolar protein sorting-35 (vps35). Replacement of 64ILV(X)4(L/V)RL74S with 64AAA(X)4(L/V)RL74S or with 64ILV(X)4AEA74A significantly reduces the binding efficiency of PTS-α to either EHD1 or EHD4 in GST pull-down assay and far western blotting assay. In addition, replacement of 218ILV(X)4VRL228S with 218AAA(X)4AEA228A decreases the binding ability of vps35 to EHD4 in far western blotting assay. Overexpression of the PTS-β in 293 T cells attenuated the endocytic trafficking of transferrin. However, this attenuation of transferrin in endocytic trafficking was disrupted when 293 T cells overexpressed the mutant PTS-β with a defective EHD-binding motif, suggesting that PTS-β can regulate the endocytic recycling via associating with EHD1 or EHD4.
Collapse
Affiliation(s)
- Kuang-Yung Huang
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
| | - Hui-Chun Yu
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Hsien-Yu Huang Tseng
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Jyun-Jie Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Ying Lin
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pin-Chen Chen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Ya-Ting Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pei-Rong Chung
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Hsiao-Kuei Tsai
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Si-Ru Zhou
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Lin Wang
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Ning-Sheng Lai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ta-Hsien Lin
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Sec. 2, Linong St., Taipei 11221, Taiwan
| | - Hsien-Bin Huang
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| |
Collapse
|
2
|
Pisanu C, Congiu D, Meloni A, Paribello P, Patrinos GP, Severino G, Ardau R, Chillotti C, Manchia M, Squassina A. Dissecting the genetic overlap between severe mental disorders and markers of cellular aging: Identification of pleiotropic genes and druggable targets. Neuropsychopharmacology 2024; 49:1033-1041. [PMID: 38402365 PMCID: PMC11039620 DOI: 10.1038/s41386-024-01822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/17/2024] [Accepted: 02/04/2024] [Indexed: 02/26/2024]
Abstract
Patients with severe mental disorders such as bipolar disorder (BD), schizophrenia (SCZ) and major depressive disorder (MDD) show a substantial reduction in life expectancy, increased incidence of comorbid medical conditions commonly observed with advanced age and alterations of aging hallmarks. While severe mental disorders are heritable, the extent to which genetic predisposition might contribute to accelerated cellular aging is not known. We used bivariate causal mixture models to quantify the trait-specific and shared architecture of mental disorders and 2 aging hallmarks (leukocyte telomere length [LTL] and mitochondrial DNA copy number), and the conjunctional false discovery rate method to detect shared genetic loci. We integrated gene expression data from brain regions from GTEx and used different tools to functionally annotate identified loci and investigate their druggability. Aging hallmarks showed low polygenicity compared with severe mental disorders. We observed a significant negative global genetic correlation between MDD and LTL (rg = -0.14, p = 6.5E-10), and no significant results for other severe mental disorders or for mtDNA-cn. However, conditional QQ plots and bivariate causal mixture models pointed to significant pleiotropy among all severe mental disorders and aging hallmarks. We identified genetic variants significantly shared between LTL and BD (n = 17), SCZ (n = 55) or MDD (n = 19), or mtDNA-cn and BD (n = 4), SCZ (n = 12) or MDD (n = 1), with mixed direction of effects. The exonic rs7909129 variant in the SORCS3 gene, encoding a member of the retromer complex involved in protein trafficking and intracellular/intercellular signaling, was associated with shorter LTL and increased predisposition to all severe mental disorders. Genetic variants underlying risk of SCZ or MDD and shorter LTL modulate expression of several druggable genes in different brain regions. Genistein, a phytoestrogen with anti-inflammatory and antioxidant effects, was an upstream regulator of 2 genes modulated by variants associated with risk of MDD and shorter LTL. While our results suggest that shared heritability might play a limited role in contributing to accelerated cellular aging in severe mental disorders, we identified shared genetic determinants and prioritized different druggable targets and compounds.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Anna Meloni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, School of Health Sciences, Department of Pharmacy, University of Patras, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al‑Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al‑Ain, Abu Dhabi, UAE
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
3
|
Zhu Y, Saribas AS, Liu J, Lin Y, Bodnar B, Zhao R, Guo Q, Ting J, Wei Z, Ellis A, Li F, Wang X, Yang X, Wang H, Ho WZ, Yang L, Hu W. Protein expression/secretion boost by a novel unique 21-mer cis-regulatory motif (Exin21) via mRNA stabilization. Mol Ther 2023; 31:1136-1158. [PMID: 36793212 PMCID: PMC9927791 DOI: 10.1016/j.ymthe.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/24/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Boosting protein production is invaluable in both industrial and academic applications. We discovered a novel expression-increasing 21-mer cis-regulatory motif (Exin21) that inserts between SARS-CoV-2 envelope (E) protein-encoding sequence and luciferase reporter gene. This unique Exin21 (CAACCGCGGTTCGCGGCCGCT), encoding a heptapeptide (QPRFAAA, designated as Qα), significantly (34-fold on average) boosted E production. Both synonymous and nonsynonymous mutations within Exin21 diminished its boosting capability, indicating the exclusive composition and order of 21 nucleotides. Further investigations demonstrated that Exin21/Qα addition could boost the production of multiple SARS-CoV-2 structural proteins (S, M, and N) and accessory proteins (NSP2, NSP16, and ORF3), and host cellular gene products such as IL-2, IFN-γ, ACE2, and NIBP. Exin21/Qα enhanced the packaging yield of S-containing pseudoviruses and standard lentivirus. Exin21/Qα addition on the heavy and light chains of human anti-SARS-CoV monoclonal antibody robustly increased antibody production. The extent of such boosting varied with protein types, cellular density/function, transfection efficiency, reporter dosage, secretion signaling, and 2A-mediated auto-cleaving efficiency. Mechanistically, Exin21/Qα increased mRNA synthesis/stability, and facilitated protein expression and secretion. These findings indicate that Exin21/Qα has the potential to be used as a universal booster for protein production, which is of importance for biomedicine research and development of bioproducts, drugs, and vaccines.
Collapse
Affiliation(s)
- Yuanjun Zhu
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - A. Sami Saribas
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Jinbiao Liu
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yuan Lin
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Brittany Bodnar
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ruotong Zhao
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Qian Guo
- Department of Medical Genetics & Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Julia Ting
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Zhengyu Wei
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Aidan Ellis
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Fang Li
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA,Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Wen-Zhe Ho
- Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ling Yang
- Department of Medical Genetics & Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Wenhui Hu
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
4
|
Cheung JP, Tubbs JD, Sham PC. Extended gene set analysis of human neuro-psychiatric traits shows enrichment in brain-expressed human accelerated regions across development. Schizophr Res 2022; 246:148-155. [PMID: 35779326 DOI: 10.1016/j.schres.2022.06.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 04/25/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022]
Abstract
Human neuropsychiatric disorders are associated with genetic and environmental factors affecting the brain, which has been subjected to strong evolutionary pressures resulting in an enlarged cerebral cortex and improved cognitive performance. Thus, genes involved in human brain evolution may also play a role in neuropsychiatric disorders. We test whether genes associated with 7 neuropsychiatric phenotypes are enriched in genomic regions that have experienced rapid changes in human evolution (HARs) and importantly, whether HAR status interacts with developmental brain expression to predict associated genes. We used the most recent publicly available GWAS and gene expression data to test for enrichment of HARs, brain expression, and their interaction. These revealed significant interactions between HAR status and whole-brain expression across developmental stages, indicating that the relationship between brain expression and association with schizophrenia and intelligence is stronger among HAR than non-HAR genes. Follow-up regional analyses indicated that predicted HAR-expression interaction effects may vary substantially across regions and developmental stages. Although depression indicated significant enrichment of HAR genes, little support was found for HAR enrichment among bipolar, autism, ADHD, or Alzheimer's associated genes. Our results indicate that intelligence, schizophrenia, and depression-associated genes are enriched for those involved in the evolution of the human brain. These findings highlight promising candidates for follow-up study and considerations for novel drug development, but also caution careful assessment of the translational ability of animal models for studying neuropsychiatric traits in the context of HARs, and the importance of using humanized animal models or human-derived tissues when researching these traits.
Collapse
Affiliation(s)
- Justin P Cheung
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Justin D Tubbs
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China; Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
5
|
Wang H, Qi W, Zou C, Xie Z, Zhang M, Naito MG, Mifflin L, Liu Z, Najafov A, Pan H, Shan B, Li Y, Zhu ZJ, Yuan J. NEK1-mediated retromer trafficking promotes blood-brain barrier integrity by regulating glucose metabolism and RIPK1 activation. Nat Commun 2021; 12:4826. [PMID: 34376696 PMCID: PMC8355301 DOI: 10.1038/s41467-021-25157-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022] Open
Abstract
Loss-of-function mutations in NEK1 gene, which encodes a serine/threonine kinase, are involved in human developmental disorders and ALS. Here we show that NEK1 regulates retromer-mediated endosomal trafficking by phosphorylating VPS26B. NEK1 deficiency disrupts endosomal trafficking of plasma membrane proteins and cerebral proteome homeostasis to promote mitochondrial and lysosomal dysfunction and aggregation of α-synuclein. The metabolic and proteomic defects of NEK1 deficiency disrupts the integrity of blood-brain barrier (BBB) by promoting lysosomal degradation of A20, a key modulator of RIPK1, thus sensitizing cerebrovascular endothelial cells to RIPK1-dependent apoptosis and necroptosis. Genetic inactivation of RIPK1 or metabolic rescue with ketogenic diet can prevent postnatal lethality and BBB damage in NEK1 deficient mice. Inhibition of RIPK1 reduces neuroinflammation and aggregation of α-synuclein in the brains of NEK1 deficient mice. Our study identifies a molecular mechanism by which retromer trafficking and metabolism regulates cerebrovascular integrity, cerebral proteome homeostasis and RIPK1-mediated neuroinflammation.
Collapse
Affiliation(s)
- Huibing Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Weiwei Qi
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Chengyu Zou
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Zhangdan Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | | | - Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Zhen Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
6
|
Unveiling the cryo-EM structure of retromer. Biochem Soc Trans 2021; 48:2261-2272. [PMID: 33125482 DOI: 10.1042/bst20200552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/29/2022]
Abstract
Retromer (VPS26/VPS35/VPS29) is a highly conserved eukaryotic protein complex that localizes to endosomes to sort transmembrane protein cargoes into vesicles and elongated tubules. Retromer mediates retrieval pathways from endosomes to the trans-Golgi network in all eukaryotes and further facilitates recycling pathways to the plasma membrane in metazoans. In cells, retromer engages multiple partners to orchestrate the formation of tubulovesicular structures, including sorting nexin (SNX) proteins, cargo adaptors, GTPases, regulators, and actin remodeling proteins. Retromer-mediated pathways are especially important for sorting cargoes required for neuronal maintenance, which links retromer loss or mutations to multiple human brain diseases and disorders. Structural and biochemical studies have long contributed to the understanding of retromer biology, but recent advances in cryo-electron microscopy and cryo-electron tomography have further uncovered exciting new snapshots of reconstituted retromer structures. These new structures reveal retromer assembles into an arch-shaped scaffold and suggest the scaffold may be flexible and adaptable in cells. Interactions with cargo adaptors, particularly SNXs, likely orient the scaffold with respect to phosphatidylinositol-3-phosphate (PtdIns3P)-enriched membranes. Pharmacological small molecule chaperones have further been shown to stabilize retromer in cultured cell and mouse models, but mechanisms by which these molecules bind remain unknown. This review will emphasize recent structural and biophysical advances in understanding retromer structure as the field moves towards a molecular view of retromer assembly and regulation on membranes.
Collapse
|
7
|
Wang X, Huang K, Yang F, Chen D, Cai S, Huang L. Association between structural brain features and gene expression by weighted gene co-expression network analysis in conversion from MCI to AD. Behav Brain Res 2021; 410:113330. [PMID: 33940051 DOI: 10.1016/j.bbr.2021.113330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease. Mild cognitive impairment (MCI) represents a state of cognitive function between normal cognition and dementia. Longitudinal studies showed that some MCI patients remained in a state of MCI, and some developed AD. The reason for these different conversions from MCI remains to be investigated. 180 MCI participants were followed for eight years. 143 MCI patients maintained the MCI state (MCI_S), and the remaining thirty-seven MCI patients were re-evaluated as having AD (MCI_AD). We obtained 1,036 structural brain characteristics and 15,481 gene expression values from the 180 MCI participants and applied weighted gene co-expression network analysis (WGCNA) to explore the relationship between structural brain features and gene expression. Regulating mediator effect analysis was employed to explore the relationships among gene expression, brain region measurements and clinical phenotypes. We found that 60 genes from the MCI_S group and 18 genes from the MCI_AD group respectively had the most significant correlations with left paracentral lobule and sulcus (L.PTS) and right subparietal sulcus (R.SubPS) thickness; CTCF, UQCR11 and WDR5B were the mutual genes between the two groups. The expression of CTCF gene and clinical score are completely mediated by L.PTS thickness, and the UQCR11 and WDR5B gene expression levels significantly regulate the mediating effect pathway. In conclusion, the factors affecting the different conversions from MCI are closely related to L.PTS thickness and the CTCF, UQCR11 and WDR5B gene expression levels. Our results add a theoretical foundation of imaging genetics for conversion from MCI to AD.
Collapse
Affiliation(s)
- Xuwen Wang
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China
| | - Kexin Huang
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China
| | - Fan Yang
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China
| | - Dihun Chen
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China
| | - Suping Cai
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China.
| | - Liyu Huang
- School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, 710071, PR China.
| |
Collapse
|
8
|
Chandra M, Kendall AK, Jackson LP. Toward Understanding the Molecular Role of SNX27/Retromer in Human Health and Disease. Front Cell Dev Biol 2021; 9:642378. [PMID: 33937239 PMCID: PMC8083963 DOI: 10.3389/fcell.2021.642378] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 11/30/2022] Open
Abstract
Aberrations in membrane trafficking pathways have profound effects in cellular dynamics of cellular sorting processes and can drive severe physiological outcomes. Sorting nexin 27 (SNX27) is a metazoan-specific sorting nexin protein from the PX-FERM domain family and is required for endosomal recycling of many important transmembrane receptors. Multiple studies have shown SNX27-mediated recycling requires association with retromer, one of the best-known regulators of endosomal trafficking. SNX27/retromer downregulation is strongly linked to Down's Syndrome (DS) via glutamate receptor dysfunction and to Alzheimer's Disease (AD) through increased intracellular production of amyloid peptides from amyloid precursor protein (APP) breakdown. SNX27 is further linked to addiction via its role in potassium channel trafficking, and its over-expression is linked to tumorigenesis, cancer progression, and metastasis. Thus, the correct sorting of multiple receptors by SNX27/retromer is vital for normal cellular function to prevent human diseases. The role of SNX27 in regulating cargo recycling from endosomes to the cell surface is firmly established, but how SNX27 assembles with retromer to generate tubulovesicular carriers remains elusive. Whether SNX27/retromer may be a putative therapeutic target to prevent neurodegenerative disease is now an emerging area of study. This review will provide an update on our molecular understanding of endosomal trafficking events mediated by the SNX27/retromer complex on endosomes.
Collapse
Affiliation(s)
- Mintu Chandra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | - Amy K. Kendall
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | - Lauren P. Jackson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
9
|
Bodnar B, DeGruttola A, Zhu Y, Lin Y, Zhang Y, Mo X, Hu W. Emerging role of NIK/IKK2-binding protein (NIBP)/trafficking protein particle complex 9 (TRAPPC9) in nervous system diseases. Transl Res 2020; 224:55-70. [PMID: 32434006 PMCID: PMC7442628 DOI: 10.1016/j.trsl.2020.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/02/2020] [Accepted: 05/05/2020] [Indexed: 02/05/2023]
Abstract
NFκB signaling and protein trafficking network play important roles in various biological and pathological processes. NIK-and-IKK2-binding protein (NIBP), also known as trafficking protein particle complex 9 (TRAPPC9), is a prototype member of a novel protein family, and has been shown to regulate both NFκB signaling pathway and protein transport/trafficking. NIBP is extensively expressed in the nervous system and plays an important role in regulating neurogenesis and neuronal differentiation. NIBP/TRAPPC9 mutations have been linked to an autosomal recessive intellectual disability syndrome, called NIBP Syndrome, which is characterized by nonsyndromic autosomal recessive intellectual disability along with other symptoms such as obesity, microcephaly, and facial dysmorphia. As more cases of NIBP Syndrome are identified, new light is being shed on the role of NIBP/TRAPPC9 in the central nervous system developments and diseases. NIBP is also involved in the enteric nervous system. This review will highlight the importance of NIBP/TRAPPC9 in central and enteric nervous system diseases, and the established possible mechanisms for developing a potential therapeutic.
Collapse
Affiliation(s)
- Brittany Bodnar
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; MD/PhD and Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Arianna DeGruttola
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; MD/PhD and Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Yuanjun Zhu
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, China
| | - Yuan Lin
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Yonggang Zhang
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Wenhui Hu
- Center for Metabolic Disease Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; MD/PhD and Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
10
|
Endosomal Trafficking in Alzheimer's Disease, Parkinson's Disease, and Neuronal Ceroid Lipofuscinosis. Mol Cell Biol 2020; 40:MCB.00262-20. [PMID: 32690545 DOI: 10.1128/mcb.00262-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuronal ceroid lipofuscinosis (NCL) is one of the most prevalent neurodegenerative disorders of early life, Parkinson's disease (PD) is the most common neurodegenerative disorder of midlife, while Alzheimer's disease (AD) is the most common neurodegenerative disorder of late life. While they are phenotypically distinct, recent studies suggest that they share a biological pathway, retromer-dependent endosomal trafficking. A retromer is a multimodular protein assembly critical for sorting and trafficking cargo out of the endosome. As a lysosomal storage disease, all 13 of NCL's causative genes affect endolysosomal function, and at least four have been directly linked to retromer. PD has several known causative genes, with one directly linked to retromer and others causing endolysosomal dysfunction. AD has over 25 causative genes/risk factors, with several of them linked to retromer or endosomal trafficking dysfunction. In this article, we summarize the emerging evidence on the association of genes causing NCL with retromer function and endosomal trafficking, review the recent evidence linking NCL genes to AD, and discuss how NCL, AD, and PD converge on a shared molecular pathway. We also discuss this pathway's role in microglia and neurons, cell populations which are critical to proper brain homeostasis and whose dysfunction plays a key role in neurodegeneration.
Collapse
|
11
|
Wang M, Wang S, Li Y, Cai G, Cao M, Li L. Integrated analysis and network pharmacology approaches to explore key genes of Xingnaojing for treatment of Alzheimer's disease. Brain Behav 2020; 10:e01610. [PMID: 32304290 PMCID: PMC7303382 DOI: 10.1002/brb3.1610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), as a neurodegenerative condition, is one of the leading causes of dementia. Our study aims to explore the key genes of Xingnaojing (XNJ) for treatment of AD by integrated microarray analysis and network pharmacology. METHODS The differentially expressed genes (DEGs) were identified in AD compared with normal control. According to these DEGs, we performed the functional annotation, protein-protein interaction (PPI) network construction. The network pharmacology was used to explore the potential targets of XNJ in the treatment of AD. The expression level of selected candidate genes was validated by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS A total of 1,424 DEGs (620 genes were upregulated and 804 genes were downregulated) between AD and normal control were obtained. The functional annotation results displayed that neuroactive ligand-receptor interaction, regulation of actin cytoskeleton, Estrogen signaling pathway and notch signaling pathway were significantly enriched pathways in AD. Comparing the target genes of four active ingredients, a total of 16 shared genes were found. Among which, HTR2A and ADRA2A were also enriched in pathway of neuroactive ligand-receptor interaction. The expression of 4 DEGs (SORCS3, HTR2A, NEFL, and TAC1) was validated by qRT-PCR. Except for TAC1, the other 3 DEGs in AD were consistent with our integrated analysis. CONCLUSIONS The results of this study may provide novel insights into the molecular mechanisms of AD and indicate potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Meixia Wang
- Department of PharmaceuticalAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Shouyong Wang
- Medication Procurement OfficeAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Yong Li
- EICUAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Gaomei Cai
- Department of Neurology WardAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Min Cao
- Continuing Education OfficeAffiliated Hospital of Jining Medical UniversityJiningChina
| | - Lanfang Li
- Department of Clinical PharmacyAffiliated Hospital of Jining Medical UniversityJiningChina
| |
Collapse
|
12
|
Chen P, Xu L, Zhang J, Cai X, Yang Y, Yu J, Qiu J, Ge J, Yu K, Zhuang J. Up-Regulation of SorCS1, an Important Sorting Receptor, in the Retina of a Form-Deprivation Rat Model. Cell Mol Neurobiol 2020; 40:395-405. [PMID: 31605284 PMCID: PMC11449034 DOI: 10.1007/s10571-019-00740-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
Visually guided regulation is a sophisticated and active process, whereby sensory input helps to shape ocular development. Here, we sought to investigate the potential involvement of SorCS1, an important protein in synaptic transmission in neuron, in retinal development. A form-deprivation (FD) rat model was established. Ocular variations induced by FD were examined, including changes to eye axial length and retinal thickness. Scotopic electroretinogram (ERG) was used to examine retinal function. RD-PCR assays were screened for differentially expressed genes in FD rat eyes. Immunofluorescence staining identified the expression pattern and localization of SorCS1 in rat retina, with or without FD treatment. Additionally, primary retinal neural cells were cultured and incubated with or without a light-dark cycle, and western blot and real-time PCR assays were used to examine the expression of SorCS1. Retinal neural cells were treated with recombinant SorCS1 (h-SorCS1) coated with beads in serum-free conditions to test for effects on cellular physiology and expression of neurotransmitters involved in visual development. To monitor cell viability, a CCK8 assay was employed. Our data demonstrated that FD led to ocular axial elongation and retinal thinning. ERG tests showed FD impaired electrophysiological function in rat. An age-related expression pattern of SorCS1 was observed in the rat retina, and SorCS1 was significantly up-regulated in the FD rat retina. In addition, in vitro evidence suggested a strong correlation between light exposure and SorCS1 expression. Furthermore, treatment of retinal neural cells with h-SorCS1-beads promoted cell viability, neurite outgrowth, and up-regulation of inhibitory neurotransmitter expression, which implies that over-expression of SorCS1 may cause abnormal retinal development. Our findings suggest that SorCS1 is involved in the physiological processes of light/visually guided ocular growth.
Collapse
Affiliation(s)
- Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Lijun Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiaoxiao Cai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jingzhi Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Malik AR, Lips J, Gorniak-Walas M, Broekaart DWM, Asaro A, Kuffner MTC, Hoffmann CJ, Kikhia M, Dopatka M, Boehm-Sturm P, Mueller S, Dirnagl U, Aronica E, Harms C, Willnow TE. SorCS2 facilitates release of endostatin from astrocytes and controls post-stroke angiogenesis. Glia 2020; 68:1304-1316. [PMID: 31898841 DOI: 10.1002/glia.23778] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/26/2023]
Abstract
SorCS2 is an intracellular sorting receptor of the VPS10P domain receptor gene family recently implicated in oxidative stress response. Here, we interrogated the relevance of stress-related activities of SorCS2 in the brain by exploring its role in ischemic stroke in mouse models and in patients. Although primarily seen in neurons in the healthy brain, expression of SorCS2 was massively induced in astrocytes surrounding the ischemic core in mice following stroke. Post-stroke induction was likely a result of increased levels of transforming growth factor β1 in damaged brain tissue, inducing Sorcs2 gene transcription in astrocytes but not neurons. Induced astrocytic expression of SorCS2 was also seen in stroke patients, substantiating the clinical relevance of this observation. In astrocytes in vitro and in the mouse brain in vivo, SorCS2 specifically controlled release of endostatin, a factor linked to post-stroke angiogenesis. The ability of astrocytes to release endostatin acutely after stroke was lost in mice deficient for SorCS2, resulting in a blunted endostatin response which coincided with impaired vascularization of the ischemic brain. Our findings identified activated astrocytes as a source for endostatin in modulation of post-stroke angiogenesis, and the importance of the sorting receptor SorCS2 in this brain stress response.
Collapse
Affiliation(s)
- Anna R Malik
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.,Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Janet Lips
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany
| | | | - Diede W M Broekaart
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Antonino Asaro
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Melanie T C Kuffner
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Christian J Hoffmann
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Majed Kikhia
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Monika Dopatka
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany.,German Centre for Neurodegenerative Diseases, Berlin, Germany
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Christoph Harms
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany
| | | |
Collapse
|
14
|
Montcouquiol M, Kelley MW. Development and Patterning of the Cochlea: From Convergent Extension to Planar Polarity. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a033266. [PMID: 30617059 DOI: 10.1101/cshperspect.a033266] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Within the mammalian cochlea, sensory hair cells and supporting cells are aligned in curvilinear rows that extend along the length of the tonotopic axis. In addition, all of the cells within the epithelium are uniformly polarized across the orthogonal neural-abneural axis. Finally, each hair cell is intrinsically polarized as revealed by the presence of an asymmetrically shaped and apically localized stereociliary bundle. It has been known for some time that many of the developmental processes that regulate these patterning events are mediated, to some extent, by the core planar cell polarity (PCP) pathway. This article will review more recent work demonstrating how components of the PCP pathway interact with cytoskeletal motor proteins to regulate cochlear outgrowth. Finally, a signaling pathway originally identified for its role in asymmetric cell divisions has recently been shown to mediate several aspects of intrinsic hair cell polarity, including kinocilia migration, bundle shape, and elongation.
Collapse
Affiliation(s)
- Mireille Montcouquiol
- INSERM, Neurocentre Magendie, U1215, F-33077 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33077 Bordeaux, France
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
15
|
Chaves G, Stanley J, Pourmand N. Mutant Huntingtin Affects Diabetes and Alzheimer's Markers in Human and Cell Models of Huntington's Disease. Cells 2019; 8:E962. [PMID: 31450785 PMCID: PMC6769852 DOI: 10.3390/cells8090962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
A higher incidence of diabetes was observed among family members of individuals affected by Huntington's Disease with no follow-up studies investigating the genetic nature of the observation. Using a genome-wide association study (GWAS), RNA sequencing (RNA-Seq) analysis and western blotting of Rattus norvegicus and human, we were able to identify that the gene family of sortilin receptors was affected in Huntington's Disease patients. We observed that less than 5% of SNPs were of statistical significance and that sortilins and HLA/MHC gene expression or SNPs were associated with mutant huntingtin (mHTT). These results suggest that ST14A cells derived from R. norvegicus are a reliable model of HD, since sortilins were identified through analysis of the transcriptome in these cells. These findings help highlight the genes involved in mechanisms targeted by diabetes drugs, such as glucose transporters as well as proteins controlling insulin release related to mHTT. To the best of our knowledge, this is the first GWAS using RNA-Seq data from both ST14A rat HD cell model and human Huntington's Disease.
Collapse
Affiliation(s)
- Gepoliano Chaves
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA 95064, USA
| | - John Stanley
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA 95064, USA
| | - Nader Pourmand
- Department of Biomolecular Engineering, University of California, Santa Cruz, CA 95064, USA.
| |
Collapse
|
16
|
Velie BD, Lillie M, Fegraeus KJ, Rosengren MK, Solé M, Wiklund M, Ihler CF, Strand E, Lindgren G. Exploring the genetics of trotting racing ability in horses using a unique Nordic horse model. BMC Genomics 2019; 20:104. [PMID: 30717660 PMCID: PMC6360714 DOI: 10.1186/s12864-019-5484-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/28/2019] [Indexed: 01/07/2023] Open
Abstract
Background Horses have been strongly selected for speed, strength, and endurance-exercise traits since the onset of domestication. As a result, highly specialized horse breeds have developed with many modern horse breeds often representing closed populations with high phenotypic and genetic uniformity. However, a great deal of variation still exists between breeds, making the horse particularly well suited for genetic studies of athleticism. To identify genomic regions associated with athleticism as it pertains to trotting racing ability in the horse, the current study applies a pooled sequence analysis approach using a unique Nordic horse model. Results Pooled sequence data from three Nordic horse populations were used for FST analysis. After strict filtering, FST analysis yielded 580 differentiated regions for trotting racing ability. Candidate regions on equine chromosomes 7 and 11 contained the largest number of SNPs (n = 214 and 147, respectively). GO analyses identified multiple genes related to intelligence, energy metabolism, and skeletal development as potential candidate genes. However, only one candidate region for trotting racing ability overlapped a known racing ability QTL. Conclusions Not unexpected for genomic investigations of complex traits, the current study identified hundreds of candidate regions contributing to trotting racing ability in the horse. Likely resulting from the cumulative effects of many variants across the genome, racing ability continues to demonstrate its polygenic nature with candidate regions implicating genes influencing both musculature and neurological development. Electronic supplementary material The online version of this article (10.1186/s12864-019-5484-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brandon D Velie
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden. .,School of Life and Environmental Sciences, University of Sydney, Sydney, Australia.
| | - Mette Lillie
- Department of Biological and Environmental Sciences, University of Gothenburg, Gothenburg, Sweden.,Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
| | - Kim Jäderkvist Fegraeus
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Maria K Rosengren
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Marina Solé
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Maja Wiklund
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Carl-Fredrik Ihler
- Department of Companion Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Eric Strand
- Department of Companion Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Gabriella Lindgren
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden.,Livestock Genetics, Department of Biosystems, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
The effects of rotenone on TH, BDNF and BDNF-related proteins in the brain and periphery: Relevance to early Parkinson's disease. J Chem Neuroanat 2019; 97:23-32. [PMID: 30690135 DOI: 10.1016/j.jchemneu.2019.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/19/2018] [Accepted: 01/17/2019] [Indexed: 12/18/2022]
Abstract
Loss of dopaminergic neurons in the substantia nigra (SN) is one of the pathological hallmarks in Parkinson's disease (PD). This neuron loss is accompanied by reduced protein and activity levels of tyrosine hydroxylase (TH), the rate-limiting enzyme of catecholamine synthesis. Reduced nigral brain-derived neurotrophic factor (BDNF) has been postulated to contribute to the loss of nigral dopaminergic neurons in PD by causing a lack of trophic support. Prior to this nigral cell loss many patients develop non-motor symptoms such as hyposmia, constipation and orthostatic hypotension. We investigated how TH, BDNF and BDNF related receptors are altered in the SN, olfactory bulb, adrenal glands and colon (which are known to be affected in PD) using rotenone-treated rats. Rotenone was administered to Sprague-Dawley rats at a dose of 2.75 mg/kg, 5 days/week for 4 weeks, via intraperitoneal injections. Rats underwent behavioural testing, and tissues were collected for western blot and ELISA analysis. This rotenone treatment induced reduced rears and distance travelled in the rearing and open field test, respectively but caused no impairments in forced movement (rotarod test). The SN had changes consistent with a pro-apoptotic state, such as increased proBDNF but no change in TH; whereas, the colon had significantly reduced TH and increased sortilin. Thus, our results indicate further investigation is warranted for this rotenone-dosing paradigm's capacity for reproducing the early stage of PD, as we observed impairments in voluntary movement and pathology in the colon without overt motor symptoms or nigral dopaminergic loss.
Collapse
|
18
|
Sun M, Zhang H. Par3 and aPKC regulate BACE1 endosome-to-TGN trafficking through PACS1. Neurobiol Aging 2017; 60:129-140. [PMID: 28946017 PMCID: PMC5653456 DOI: 10.1016/j.neurobiolaging.2017.08.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/11/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
The cleavage of amyloid precursor protein (APP) by β-site APP cleaving enzyme 1 (BACE1) is the rate-limiting step in beta amyloid generation during Alzheimer's disease (AD) pathogenesis. In AD brains, BACE1 is abnormally accumulated in endocytic compartments, where the acidic pH is optimal for its activity. However, mechanisms regulating the endosome-to-trans-Golgi network (TGN) retrieval of BACE1 remain unclear. Here, we show that partitioning defective 3 (Par3) facilitates BACE1 retrograde trafficking from endosomes to the TGN. Par3 functions through aPKC-mediated phosphorylation of BACE1 on Ser498, which in turn promotes the interaction between BACE1 and phosphofurin acidic cluster sorting protein 1 and facilitates the retrograde trafficking of BACE1 to the TGN. In human AD brains, there is a significant decrease in Ser498 phosphorylation of BACE1 suggesting that defective phosphorylation-dependent retrograde transport of BACE1 is important in AD pathogenesis. Together, our studies provide mechanistic insight into a novel role for Par3 and aPKC in regulating the retrograde endosome-to-TGN trafficking of BACE1 and shed light on the mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Miao Sun
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
19
|
Leloup N, Lössl P, Meijer DH, Brennich M, Heck AJR, Thies-Weesie DME, Janssen BJC. Low pH-induced conformational change and dimerization of sortilin triggers endocytosed ligand release. Nat Commun 2017; 8:1708. [PMID: 29167428 PMCID: PMC5700061 DOI: 10.1038/s41467-017-01485-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 09/19/2017] [Indexed: 11/24/2022] Open
Abstract
Low pH-induced ligand release and receptor recycling are important steps for endocytosis. The transmembrane protein sortilin, a β-propeller containing endocytosis receptor, internalizes a diverse set of ligands with roles in cell differentiation and homeostasis. The molecular mechanisms of pH-mediated ligand release and sortilin recycling are unresolved. Here we present crystal structures that show the sortilin luminal segment (s-sortilin) undergoes a conformational change and dimerizes at low pH. The conformational change, within all three sortilin luminal domains, provides an altered surface and the dimers sterically shield a large interface while bringing the two s-sortilin C-termini into close proximity. Biophysical and cell-based assays show that members of two different ligand families, (pro)neurotrophins and neurotensin, preferentially bind the sortilin monomer. This indicates that sortilin dimerization and conformational change discharges ligands and triggers recycling. More generally, this work may reveal a double mechanism for low pH-induced ligand release by endocytosis receptors. Sortilin is an endocytosis receptor with a luminal β-propeller domain. Here the authors present the structures of the β-propeller domain at neutral and acidic pH, which reveal that sortilin dimerises and undergoes conformational changes at low pH and further propose a model for low pH-induced ligand release by endocytosis receptors.
Collapse
Affiliation(s)
- Nadia Leloup
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Philip Lössl
- Biomolecular Mass Spectrometry & Proteomics and Netherlands Proteomics Center, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Dimphna H Meijer
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Martha Brennich
- European Molecular Biology Laboratory, Grenoble Outstation, Grenoble, 38000, France
| | - Albert J R Heck
- Biomolecular Mass Spectrometry & Proteomics and Netherlands Proteomics Center, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Dominique M E Thies-Weesie
- Van't Hoff Laboratory for Physical and Colloid Chemistry, Debye Institute for Nanomaterials Science, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Bert J C Janssen
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Science, Utrecht University, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
20
|
McMillan KJ, Korswagen HC, Cullen PJ. The emerging role of retromer in neuroprotection. Curr Opin Cell Biol 2017; 47:72-82. [PMID: 28399507 PMCID: PMC5677836 DOI: 10.1016/j.ceb.2017.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 11/26/2022]
Abstract
Efficient sorting and transportation of integral membrane proteins, such as ion channels, nutrient transporters, signalling receptors, cell-cell and cell-matrix adhesion molecules is essential for the function of cellular organelles and hence organism development and physiology. Retromer is a master controller of integral membrane protein sorting and transport through one of the major sorting station within eukaryotic cells, the endosomal network. Subtle de-regulation of retromer is an emerging theme in the pathoetiology of Parkinson's disease. Here we summarise recent advances in defining the neuroprotective role of retromer and how its de-regulation may contribute to Parkinson's disease by interfering with: lysosomal health and protein degradation, association with accessory proteins including the WASH complex and mitochondrial health.
Collapse
Affiliation(s)
- Kirsty J McMillan
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD, UK
| | - Hendrick C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD, UK.
| |
Collapse
|
21
|
Martinez A, Lectez B, Ramirez J, Popp O, Sutherland JD, Urbé S, Dittmar G, Clague MJ, Mayor U. Quantitative proteomic analysis of Parkin substrates in Drosophila neurons. Mol Neurodegener 2017; 12:29. [PMID: 28399880 PMCID: PMC5387213 DOI: 10.1186/s13024-017-0170-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/30/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Parkin (PARK2) is an E3 ubiquitin ligase that is commonly mutated in Familial Parkinson's Disease (PD). In cell culture models, Parkin is recruited to acutely depolarised mitochondria by PINK1. PINK1 activates Parkin activity leading to ubiquitination of multiple proteins, which in turn promotes clearance of mitochondria by mitophagy. Many substrates have been identified using cell culture models in combination with depolarising drugs or proteasome inhibitors, but not in more physiological settings. METHODS Here we utilized the recently introduced BioUb strategy to isolate ubiquitinated proteins in flies. Following Parkin Wild-Type (WT) and Parkin Ligase dead (LD) expression we analysed by mass spectrometry and stringent bioinformatics analysis those proteins differentially ubiquitinated to provide the first survey of steady state Parkin substrates using an in vivo model. We further used an in vivo ubiquitination assay to validate one of those substrates in SH-SY5Y cells. RESULTS We identified 35 proteins that are more prominently ubiquitinated following Parkin over-expression. These include several mitochondrial proteins and a number of endosomal trafficking regulators such as v-ATPase sub-units, Syx5/STX5, ALiX/PDCD6IP and Vps4. We also identified the retromer component, Vps35, another PD-associated gene that has recently been shown to interact genetically with parkin. Importantly, we validated Parkin-dependent ubiquitination of VPS35 in human neuroblastoma cells. CONCLUSIONS Collectively our results provide new leads to the possible physiological functions of Parkin activity that are not overtly biased by acute mitochondrial depolarisation.
Collapse
Affiliation(s)
- Aitor Martinez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,Functional Genomics Unit, CIC bioGUNE, Derio, Spain.,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Benoit Lectez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,Functional Genomics Unit, CIC bioGUNE, Derio, Spain
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Sylvie Urbé
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gunnar Dittmar
- Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Michael J Clague
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain. .,Functional Genomics Unit, CIC bioGUNE, Derio, Spain. .,Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain.
| |
Collapse
|
22
|
Forge A, Taylor RR, Dawson SJ, Lovett M, Jagger DJ. Disruption of SorCS2 reveals differences in the regulation of stereociliary bundle formation between hair cell types in the inner ear. PLoS Genet 2017; 13:e1006692. [PMID: 28346477 PMCID: PMC5386298 DOI: 10.1371/journal.pgen.1006692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/10/2017] [Accepted: 03/14/2017] [Indexed: 12/24/2022] Open
Abstract
Behavioural anomalies suggesting an inner ear disorder were observed in a colony of transgenic mice. Affected animals were profoundly deaf. Severe hair bundle defects were identified in all outer and inner hair cells (OHC, IHC) in the cochlea and in hair cells of vestibular macular organs, but hair cells in cristae were essentially unaffected. Evidence suggested the disorder was likely due to gene disruption by a randomly inserted transgene construct. Whole-genome sequencing identified interruption of the SorCS2 (Sortilin-related VPS-10 domain containing protein) locus. Real-time-qPCR demonstrated disrupted expression of SorCS2 RNA in cochlear tissue from affected mice and this was confirmed by SorCS2 immuno-labelling. In all affected hair cells, stereocilia were shorter than normal, but abnormalities of bundle morphology and organisation differed between hair cell types. Bundles on OHC were grossly misshapen with significantly fewer stereocilia than normal. However, stereocilia were organised in rows of increasing height. Bundles on IHC contained significantly more stereocilia than normal with some longer stereocilia towards the centre, or with minimal height differentials. In early postnatal mice, kinocilia (primary cilia) of IHC and of OHC were initially located towards the lateral edge of the hair cell surface but often became surrounded by stereocilia as bundle shape and apical surface contour changed. In macular organs the kinocilium was positioned in the centre of the cell surface throughout maturation. There was disruption of the signalling pathway controlling intrinsic hair cell apical asymmetry. LGN and Gαi3 were largely absent, and atypical Protein Kinase C (aPKC) lost its asymmetric distribution. The results suggest that SorCS2 plays a role upstream of the intrinsic polarity pathway and that there are differences between hair cell types in the deployment of the machinery that generates a precisely organised hair bundle.
Collapse
MESH Headings
- Age Factors
- Animals
- Gene Expression Regulation
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/pathology
- Hearing Loss/genetics
- Hearing Loss/metabolism
- Hearing Loss/physiopathology
- Immunohistochemistry
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Microscopy, Electron, Scanning
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Organ of Corti/metabolism
- Organ of Corti/physiopathology
- Organ of Corti/ultrastructure
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Stereocilia/genetics
- Stereocilia/metabolism
- Stereocilia/pathology
Collapse
Affiliation(s)
- Andrew Forge
- UCL Ear Institute, University College London, London, United Kingdom
- * E-mail:
| | - Ruth R. Taylor
- UCL Ear Institute, University College London, London, United Kingdom
| | - Sally J. Dawson
- UCL Ear Institute, University College London, London, United Kingdom
| | - Michael Lovett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Daniel J. Jagger
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
23
|
Role of the VPS35 D620N mutation in Parkinson's disease. Parkinsonism Relat Disord 2017; 36:10-18. [DOI: 10.1016/j.parkreldis.2016.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
|
24
|
Chu J, Praticò D. The retromer complex system in a transgenic mouse model of AD: influence of age. Neurobiol Aging 2017; 52:32-38. [PMID: 28110103 DOI: 10.1016/j.neurobiolaging.2016.12.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/21/2016] [Accepted: 12/27/2016] [Indexed: 10/20/2022]
Abstract
Deficiencies of the retrograde transport mediated by the retromer complex have been described in Alzheimer's disease (AD). Genetic manipulation of retromer modulates brain amyloidosis in Tg2576 mice. However, whether the complex is altered during the development of the AD-like phenotype remains unknown. In this study we assayed the expression levels of the vacuolar sorting protein 35 (VPS35), VPS26, VPS29, and its cargo proteins, cation independent mannose 6-phosphate receptor, sortilin-related receptor in brains of Tg2576 and controls at the ages of 3, 8, and 14 months. While cortex showed an age-dependent decrease in all but VPS29, levels of the same proteins in the cerebellum were unchanged at any age. Neuronal cells expressing human amyloid beta precursor protein Swedish mutant had also reduced retromer complex levels. However, incubation with a pharmacological chaperone dose-dependently restored these levels together with a reduction in amyloid beta. Our study is the first to show that in a transgenic mouse model of AD the changes in the expression levels of the retromer complex are age and region dependent, and that the complex is a viable therapeutic target since its deficiency can be restored pharmacologically by a retromer chaperone.
Collapse
Affiliation(s)
- Jin Chu
- Department of Pharmacology and Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Domenico Praticò
- Department of Pharmacology and Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Coria-Lucero CD, Golini RS, Ponce IT, Deyurka N, Anzulovich AC, Delgado SM, Navigatore-Fonzo LS. Rhythmic Bdnf and TrkB expression patterns in the prefrontal cortex are lost in aged rats. Brain Res 2016; 1653:51-58. [PMID: 27771283 DOI: 10.1016/j.brainres.2016.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 10/10/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022]
Abstract
Aging brain undergoes several changes leading to a decline in cognitive functions. Memory and learning-related genes such as Creb, Bdnf and its receptor TrkB, are expressed in different brain regions including prefrontal cortex. Those genes' proteins regulate a wide range of functions such as synaptic plasticity and long-term potentiation. In this work, our objectives were: 1) to investigate whether Creb1, Bdnf and TrkB genes display endogenous circadian expression rhythms, in the prefrontal cortex of rats maintained under constant darkness conditions; 2) to study the synchronization of those temporal patterns to the local cellular clock and 3) to evaluate the aging consequences on both cognition-related genes and activating clock transcription factor, BMAL1, rhythms. A bioinformatics analysis revealed clock-responsive (E-box) sites in regulatory regions of Creb1, Bdnf and TrkB genes. Additionally, cAMP response elements (CRE) were found in Bdnf and TrkB promoters. We observed those key cognition-related factors expression oscillates in the rat prefrontal cortex. Creb1 and TrkB mRNAs display a circadian rhythm with their highest levels occurring at the second half of the 24h period. Interestingly, the cosinor analysis revealed a 12-h rhythm of Bdnf transcript levels, with peaks occurring at the second half of the subjective day and night, respectively. As expected, the BMAL1 rhythm's acrophase precedes Creb1 and first Bdnf expression peaks. Noteworthy, Creb1, Bdnf and TrkB expression rhythms are lost in the prefrontal cortex of aged rats, probably, as consequence of the loss of BMAL1 protein circadian rhythm and altered function of the local cellular clock.
Collapse
Affiliation(s)
- Cinthia D Coria-Lucero
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Rebeca S Golini
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Ivana T Ponce
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Nicolas Deyurka
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Ana C Anzulovich
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Silvia M Delgado
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina; Laboratory of Biology Reproduction, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina
| | - Lorena S Navigatore-Fonzo
- Laboratory of Chronobiology, Multidisciplinary Institute of Biological Research-San Luis (IMIBIO-SL), National Council of Science and Technology (CONICET), National University of San Luis (UNSL)., Av Ejército de los Andes N° 950, D5700HHW, San Luis, Argentina.
| |
Collapse
|
26
|
Alzheimer disease: modeling an Aβ-centered biological network. Mol Psychiatry 2016; 21:861-71. [PMID: 27021818 DOI: 10.1038/mp.2016.38] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/15/2023]
Abstract
In genetically complex diseases, the search for missing heritability is focusing on rare variants with large effect. Thanks to next generation sequencing technologies, genome-wide characterization of these variants is now feasible in every individual. However, a lesson from current studies is that collapsing rare variants at the gene level is often insufficient to obtain a statistically significant signal in case-control studies, and that network-based analyses are an attractive complement to classical approaches. In Alzheimer disease (AD), according to the prevalent amyloid cascade hypothesis, the pathology is driven by the amyloid beta (Aβ) peptide. In past years, based on experimental studies, several hundreds of proteins have been shown to interfere with Aβ production, clearance, aggregation or toxicity. Thanks to a manual curation of the literature, we identified 335 genes/proteins involved in this biological network and classified them according to their cellular function. The complete list of genes, or its subcomponents, will be of interest in ongoing AD genetic studies.
Collapse
|
27
|
Li C, Shah SZA, Zhao D, Yang L. Role of the Retromer Complex in Neurodegenerative Diseases. Front Aging Neurosci 2016; 8:42. [PMID: 26973516 PMCID: PMC4772447 DOI: 10.3389/fnagi.2016.00042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Abstract
The retromer complex is a protein complex that plays a central role in endosomal trafficking. Retromer dysfunction has been linked to a growing number of neurological disorders. The process of intracellular trafficking and recycling is crucial for maintaining normal intracellular homeostasis, which is partly achieved through the activity of the retromer complex. The retromer complex plays a primary role in sorting endosomal cargo back to the cell surface for reuse, to the trans-Golgi network (TGN), or alternatively to specialized endomembrane compartments, in which the cargo is not subjected to lysosomal-mediated degradation. In most cases, the retromer acts as a core that interacts with associated proteins, including sorting nexin family member 27 (SNX27), members of the vacuolar protein sorting 10 (VPS10) receptor family, the major endosomal actin polymerization-promoting complex known as Wiskott-Aldrich syndrome protein and scar homolog (WASH), and other proteins. Some of the molecules carried by the retromer complex are risk factors for neurodegenerative diseases. Defects such as haplo-insufficiency or mutations in one or several units of the retromer complex lead to various pathologies. Here, we summarize the molecular architecture of the retromer complex and the roles of this system in intracellular trafficking related the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Chaosi Li
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University Beijing, China
| | - Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University Beijing, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University Beijing, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University Beijing, China
| |
Collapse
|
28
|
Vergés M. Retromer in Polarized Protein Transport. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:129-79. [PMID: 26944621 DOI: 10.1016/bs.ircmb.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Retromer is an evolutionary conserved protein complex required for endosome-to-Golgi retrieval of receptors for lysosomal hydrolases. It is constituted by a heterotrimer encoded by the vacuolar protein sorting (VPS) gene products Vps26, Vps35, and Vps29, which selects cargo, and a dimer of phosphoinositide-binding sorting nexins, which deforms the membrane. Recent progress in the mechanism of retromer assembly and functioning has strengthened the link between sorting at the endosome and cytoskeleton dynamics. Retromer is implicated in endosomal sorting of many cargos and plays an essential role in plant and animal development. Although it is best known for endosome sorting to the trans-Golgi network, it also intervenes in recycling to the plasma membrane. In polarized cells, such as epithelial cells and neurons, retromer may also be utilized for transcytosis and long-range transport. Considerable evidence implicates retromer in establishment and maintenance of cell polarity. That includes sorting of the apical polarity module Crumbs; regulation of retromer function by the basolateral polarity module Scribble; and retromer-dependent recycling of various cargoes to a certain surface domain, thus controlling polarized location and cell homeostasis. Importantly, altered retromer function has been linked to neurodegeneration, such as in Alzheimer's or Parkinson's disease. This review will underline how alterations in retromer localization and function may affect polarized protein transport and polarity establishment, thereby causing developmental defects and disease.
Collapse
Affiliation(s)
- Marcel Vergés
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Medical Sciences Department, University of Girona, Girona, Spain.
| |
Collapse
|
29
|
Hu YB, Dammer EB, Ren RJ, Wang G. The endosomal-lysosomal system: from acidification and cargo sorting to neurodegeneration. Transl Neurodegener 2015; 4:18. [PMID: 26448863 PMCID: PMC4596472 DOI: 10.1186/s40035-015-0041-1] [Citation(s) in RCA: 433] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
The endosomal-lysosomal system is made up of a set of intracellular membranous compartments that dynamically interconvert, which is comprised of early endosomes, recycling endosomes, late endosomes, and the lysosome. In addition, autophagosomes execute autophagy, which delivers intracellular contents to the lysosome. Maturation of endosomes and/or autophagosomes into a lysosome creates an unique acidic environment within the cell for proteolysis and recycling of unneeded cellular components into usable amino acids and other biomolecular building blocks. In the endocytic pathway, gradual maturation of endosomes into a lysosome and acidification of the late endosome are accompanied by vesicle trafficking, protein sorting and targeted degradation of some sorted cargo. Two opposing sorting systems are operating in these processes: the endosomal sorting complex required for transport (ESCRT) supports targeted degradation and the retromer supports retrograde retrieval of certain cargo. The endosomal-lysosomal system is emerging as a central player in a host of neurodegenerative diseases, demonstrating potential roles which are likely to be revealed in pathogenesis and for viable therapeutic strategies. Here we focus on the physiological process of endosomal-lysosomal maturation, acidification and sorting systems along the endocytic pathway, and further discuss relationships between abnormalities in the endosomal-lysosomal system and neurodegenerative diseases, especially Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Yong-Bo Hu
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Eric B Dammer
- Department of Biochemistry, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Ru-Jing Ren
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Gang Wang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
30
|
Binzer S, Stenager E, Binzer M, Kyvik KO, Hillert J, Imrell K. Genetic analysis of the isolated Faroe Islands reveals SORCS3 as a potential multiple sclerosis risk gene. Mult Scler 2015; 22:733-40. [DOI: 10.1177/1352458515602338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/21/2015] [Indexed: 11/15/2022]
Abstract
Background: In search of the missing heritability in multiple sclerosis (MS), additional approaches adding to the genetic discoveries of large genome-wide association studies are warranted. Objective: The objective of this research paper is to search for rare genetic MS risk variants in the genetically homogenous population of the isolated Faroe Islands. Methods: Twenty-nine Faroese MS cases and 28 controls were genotyped with the HumanOmniExpressExome-chip. The individuals make up 1596 pair-combinations in which we searched for identical-by-descent shared segments using the PLINK-program. Results: A segment spanning 63 SNPs with excess case-case-pair sharing was identified (0.00173 < p > 0.00212). A haplotype consisting of 42 of the 63 identified SNPs which spanned the entire the Sortilin-related vacuolar protein sorting 10 domain containing receptor 3 ( SORCS3) gene had a carrier frequency of 0.34 in cases but was not present in any controls ( p = 0.0008). Conclusion: This study revealed an oversharing in case-case-pairs of a segment spanning 63 SNPs and the entire SORCS3. While not previously associated with MS, SORCS3 appears to be important in neuronal plasticity through its binding of neurotrophin factors and involvement in glutamate homeostasis. Although additional work is needed to scrutinise the genetic effect of the SORCS3-covering haplotype, this study suggests that SORCS3 may also be important in MS pathogenesis.
Collapse
Affiliation(s)
- S Binzer
- Institute of Regional Health Research, University of Southern Denmark, Denmark/Hospital of Southern Jutland, Denmark/Odense Patient data Explorative Network (OPEN), University of Southern Denmark, Denmark/ Torshavn National Hospital, Faroe Islands
| | - E Stenager
- Institute of Regional Health Research, University of Southern Denmark, Denmark/Hospital of Southern Jutland, Denmark/ MS Clinic of Southern Jutland (Sønderborg, Esbjerg, Vejle), Department of Neurology, Denmark
| | - M Binzer
- Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - KO Kyvik
- Department of Clinical Research, University of Southern Denmark, Denmark/Odense Patient data Explorative Network (OPEN), University of Southern Denmark, Denmark
| | - J Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - K Imrell
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Abstract
The retromer complex is an important component of the endosomal protein sorting machinery and mediates protein cargoes from endosomes to the trans-Golgi network (TGN) by retrograde pathway or to the cell surface through recycling pathway. Studies show that retromer and its receptors can make amyloid precursor protein (APP)/β-site APP-cleaving enzyme 1 (BACE1) away endosomes that reduces the production of amyloid β (Aβ). And, tetramer is also found to regulate phagocytic receptors to the plasma membrane of microglia, where some phagocytic receptors take part in Aβ clearance. Therefore, disruption of retromer will increase the production of Aβ. Recently, a plausible relationship between disturbance of retromer and tauopathies is raised. Retromer dysfunction may result in decreasing the clearance of extracellular tau and the level of cathepsin D, which enables tau-induced neurotoxicity. This review article summarizes the structure and function of retromer and its role in pathogenesis of AD. In the end, retromer may provide a potential therapeutic strategy for the treatment of AD.
Collapse
|
32
|
Damseh N, Danson CM, Al-Ashhab M, Abu-Libdeh B, Gallon M, Sharma K, Yaacov B, Coulthard E, Caldwell MA, Edvardson S, Cullen PJ, Elpeleg O. A defect in the retromer accessory protein, SNX27, manifests by infantile myoclonic epilepsy and neurodegeneration. Neurogenetics 2015; 16:215-221. [PMID: 25894286 PMCID: PMC4962907 DOI: 10.1007/s10048-015-0446-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/20/2015] [Indexed: 10/23/2022]
Abstract
The composition of the neuronal cell surface dictates synaptic plasticity and thereby cognitive development. This remodeling of the synapses is governed by the endocytic network which internalize transmembrane proteins, then sort them back to the cell surface or carry them to the lysosome for degradation. The multi-protein retromer complex is central to this selection, capturing specific transmembrane proteins and remodeling the cell membrane to form isolated cargo-enriched transport carriers. We investigated a consanguineous family with four patients who presented in infancy with intractable myoclonic epilepsy and lack of psychomotor development. Using exome analysis, we identified a homozygous deleterious mutation in SNX27, which encodes sorting nexin 27, a retromer cargo adaptor. In western analysis of patient fibroblasts, the encoded mutant protein was expressed at an undetectable level when compared with a control sample. The patients' presentation and clinical course recapitulate that reported for the SNX27 knock-out mouse. Since the cargo proteins for SNX27-mediated sorting include subunits of ionotropic glutamate receptors and endosome-to-cell surface synaptic insertion of AMPA receptors is severely perturbed in SNX27(-/-) neurons, it is proposed that at least part of the neurological aberrations observed in the patients is attributed to defective sorting of ionotropic glutamate receptors. SNX27 deficiency is now added to the growing list of neurodegenerative disorders associated with retromer dysfunction.
Collapse
Affiliation(s)
- Nadirah Damseh
- Department of Pediatrics, Al-Makassed Islamic Hospital, Jerusalem, Israel
| | - Chris M. Danson
- The Henry Wellcome Integrated Signalling Laboratories, School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Motee Al-Ashhab
- Department of Pediatrics, Al-Makassed Islamic Hospital, Jerusalem, Israel
| | - Bassam Abu-Libdeh
- Department of Pediatrics, Al-Makassed Islamic Hospital, Jerusalem, Israel
| | - Matthew Gallon
- The Henry Wellcome Integrated Signalling Laboratories, School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Kanchan Sharma
- ReMemBr Group, Institute of Clinical Neurosciences, University of Bristol and North Bristol NHS Trust, Learning and Research Building, Southmead Hospital, Bristol BS10 5NB, UK
| | - Barak Yaacov
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center Jerusalem, Jerusalem, Israel
| | - Elizabeth Coulthard
- ReMemBr Group, Institute of Clinical Neurosciences, University of Bristol and North Bristol NHS Trust, Learning and Research Building, Southmead Hospital, Bristol BS10 5NB, UK
| | - Maeve A. Caldwell
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol BS1 3NY, UK
| | - Simon Edvardson
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center Jerusalem, Jerusalem, Israel
| | - Peter J. Cullen
- The Henry Wellcome Integrated Signalling Laboratories, School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center Jerusalem, Jerusalem, Israel
| |
Collapse
|
33
|
Mukadam AS, Seaman MNJ. Retromer-mediated endosomal protein sorting: The role of unstructured domains. FEBS Lett 2015; 589:2620-6. [PMID: 26072290 DOI: 10.1016/j.febslet.2015.05.052] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 05/21/2015] [Accepted: 05/26/2015] [Indexed: 12/21/2022]
Abstract
The retromer complex is a key element of the endosomal protein sorting machinery that is conserved through evolution and has been shown to play a role in diseases such as Alzheimer's disease and Parkinson's disease. Through sorting various membrane proteins (cargo), the function of retromer complex has been linked to physiological processes such as lysosome biogenesis, autophagy, down regulation of signalling receptors and cell spreading. The cargo-selective trimer of retromer recognises membrane proteins and sorts them into two distinct pathways; endosome-to-Golgi retrieval and endosome-to-cell surface recycling and additionally the cargo-selective trimer functions as a hub to recruit accessory proteins to endosomes where they may regulate and/or facilitate retromer-mediated endosomal proteins sorting. Unstructured domains present in cargo proteins or accessory factors play key roles in both these aspects of retromer function and will be discussed in this review.
Collapse
Affiliation(s)
- Aamir S Mukadam
- Cambridge Institute for Medical Research, Dept. of Clinical Biochemistry, University of Cambridge, Wellcome Trust/MRC Building, Addenbrookes Hospital, Cambridge CB2 0XY, United Kingdom
| | - Matthew N J Seaman
- Cambridge Institute for Medical Research, Dept. of Clinical Biochemistry, University of Cambridge, Wellcome Trust/MRC Building, Addenbrookes Hospital, Cambridge CB2 0XY, United Kingdom.
| |
Collapse
|