101
|
Tu Y, Zhao L, Billadeau DD, Jia D. Endosome-to-TGN Trafficking: Organelle-Vesicle and Organelle-Organelle Interactions. Front Cell Dev Biol 2020; 8:163. [PMID: 32258039 PMCID: PMC7093645 DOI: 10.3389/fcell.2020.00163] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022] Open
Abstract
Retrograde transport from endosomes to the trans-Golgi network (TGN) diverts proteins and lipids away from lysosomal degradation. It is essential for maintaining cellular homeostasis and signaling. In recent years, significant advancements have been made in understanding this classical pathway, revealing new insights into multiple steps of vesicular trafficking as well as critical roles of ER-endosome contacts for endosomal trafficking. In this review, we summarize up-to-date knowledge about this trafficking pathway, in particular, mechanisms of cargo recognition at endosomes and vesicle tethering at the TGN, and contributions of ER-endosome contacts.
Collapse
Affiliation(s)
- Yingfeng Tu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, State Key Laboratory of Biotherapy, Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, State Key Laboratory of Biotherapy, Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Daniel D. Billadeau
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, State Key Laboratory of Biotherapy, Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
102
|
Cunningham LA, Moore DJ. Endosomal sorting pathways in the pathogenesis of Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:271-306. [PMID: 32247367 PMCID: PMC7206894 DOI: 10.1016/bs.pbr.2020.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The identification of Parkinson's disease (PD)-associated genes has created a powerful platform to begin to understand and nominate pathophysiological disease mechanisms. Herein, we discuss the genetic and experimental evidence supporting endolysosomal dysfunction as a major pathway implicated in PD. Well-studied familial PD-linked gene products, including LRRK2, VPS35, and α-synuclein, demonstrate how disruption of different aspects of endolysosomal sorting pathways by disease-causing mutations may manifest into PD-like phenotypes in many disease models. Newly-identified PD-linked genes, including auxilin, synaptojanin-1 and Rab39b, as well as putative risk genes for idiopathic PD (endophilinA1, Rab29, GAK), further support endosomal sorting deficits as being central to PD. LRRK2 may represent a nexus by regulating many distinct features of endosomal sorting, potentially via phosphorylation of key endocytosis machinery (i.e., auxilin, synaptojanin-1, endoA1) and Rab GTPases (i.e., Rab29, Rab8A, Rab10) that function within these pathways. In turn, LRRK2 kinase activity is critically regulated by Rab29 at the Golgi complex and retromer-associated VPS35 at endosomes. Taken together, the known functions of PD-associated gene products, the impact of disease-linked mutations, and the emerging functional interactions between these proteins points to endosomal sorting pathways as a key point of convergence in the pathogenesis of PD.
Collapse
Affiliation(s)
- Lindsey A Cunningham
- Van Andel Institute Graduate School, Grand Rapids, MI, United States; Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States.
| |
Collapse
|
103
|
Yasa S, Modica G, Sauvageau E, Kaleem A, Hermey G, Lefrancois S. CLN3 regulates endosomal function by modulating Rab7A-effector interactions. J Cell Sci 2020; 133:jcs.234047. [PMID: 32034082 DOI: 10.1242/jcs.234047] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/22/2020] [Indexed: 01/02/2023] Open
Abstract
Mutations in CLN3 are a cause of juvenile neuronal ceroid lipofuscinosis (JNCL), also known as Batten disease. Clinical manifestations include cognitive regression, progressive loss of vision and motor function, epileptic seizures and a significantly reduced lifespan. CLN3 localizes to endosomes and lysosomes, and has been implicated in intracellular trafficking and autophagy. However, the precise molecular function of CLN3 remains to be elucidated. Previous studies showed an interaction between CLN3 and Rab7A, a small GTPase that regulates several functions at late endosomes. We confirmed this interaction in live cells and found that CLN3 is required for the efficient endosome-to-TGN trafficking of the lysosomal sorting receptors because it regulates the Rab7A interaction with retromer. In cells lacking CLN3 or expressing CLN3 harbouring a disease-causing mutation, the lysosomal sorting receptors were degraded. We also demonstrated that CLN3 is required for the Rab7A-PLEKHM1 interaction, which is required for fusion of autophagosomes to lysosomes. Overall, our data provide a molecular explanation behind phenotypes observed in JNCL and give an indication of the pathogenic mechanism behind Batten disease.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Seda Yasa
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada H7V 1B7
| | - Graziana Modica
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada H7V 1B7
| | - Etienne Sauvageau
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada H7V 1B7
| | - Abuzar Kaleem
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Stephane Lefrancois
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada H7V 1B7 .,Department of Anatomy and Cell Biology, McGill University, Montreal, Canada H3A 0C7.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, Canada H2X 3Y7
| |
Collapse
|
104
|
Yong X, Zhao L, Deng W, Sun H, Zhou X, Mao L, Hu W, Shen X, Sun Q, Billadeau DD, Xue Y, Jia D. Mechanism of cargo recognition by retromer-linked SNX-BAR proteins. PLoS Biol 2020; 18:e3000631. [PMID: 32150533 PMCID: PMC7082075 DOI: 10.1371/journal.pbio.3000631] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 03/19/2020] [Accepted: 02/21/2020] [Indexed: 12/30/2022] Open
Abstract
Endocytic recycling of internalized transmembrane proteins is essential for many important physiological processes. Recent studies have revealed that retromer-related Sorting Nexin family (SNX)–Bin/Amphiphysin/Rvs (BAR) proteins can directly recognize cargoes like cation-independent mannose 6-phosphate receptor (CI-MPR) and Insulin-like growth factor 1 receptor (IGF1R); however, it remains poorly understood how SNX-BARs select specific cargo proteins and whether they recognize additional ligands. Here, we discovered that the binding between SNX-BARs and CI-MPR or IGF1R is mediated by the phox-homology (PX) domain of SNX5 or SNX6 and a bipartite motif, termed SNX-BAR-binding motif (SBM), in the cargoes. Using this motif, we identified over 70 putative SNX-BAR ligands, many of which play critical roles in apoptosis, cell adhesion, signal transduction, or metabolite homeostasis. Remarkably, SNX-BARs could cooperate with both SNX27 and retromer in the recycling of ligands encompassing the SBM, PDZ-binding motif, or both motifs. Overall, our studies establish that SNX-BARs function as a direct cargo-selecting module for a large set of transmembrane proteins transiting the endosome, in addition to their roles in phospholipid recognition and biogenesis of tubular structures. Internalized transmembrane proteins can be recognized by specific protein complexes and diverted away from the degradation process. This study identifies a new sorting motif recognized by retromer-linked SNX-BAR proteins and reveals a large repertoire of potential cargoes recycled by the SNX-BAR proteins.
Collapse
Affiliation(s)
- Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Wankun Deng
- Department of Bioinformatics & Systems Biology, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbin Sun
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou, China
| | - Xue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Lejiao Mao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Wenfeng Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaofei Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Qingxiang Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Daniel D. Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Yu Xue
- Department of Bioinformatics & Systems Biology, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
105
|
Dhawan K, Naslavsky N, Caplan S. Sorting nexin 17 (SNX17) links endosomal sorting to Eps15 homology domain protein 1 (EHD1)-mediated fission machinery. J Biol Chem 2020; 295:3837-3850. [PMID: 32041776 DOI: 10.1074/jbc.ra119.011368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Following endocytosis, receptors that are internalized to sorting endosomes are sorted to different pathways, in part by sorting nexin (SNX) proteins. Notably, SNX17 interacts with a multitude of receptors in a sequence-specific manner to regulate their recycling. However, the mechanisms by which SNX17-labeled vesicles that contain sorted receptors bud and undergo vesicular fission from the sorting endosomes remain elusive. Recent studies suggest that a dynamin-homolog, Eps15 homology domain protein 1, catalyzes fission and releases endosome-derived vesicles for recycling to the plasma membrane. However, the mechanism by which EHD1 is coupled to various receptors and regulates their recycling remains unknown. Here we sought to characterize the mechanism by which EHD1 couples with SNX17 to regulate recycling of SNX17-interacting receptors. We hypothesized that SNX17 couples receptors to the EHD1 fission machinery in mammalian cells. Coimmunoprecipitation experiments and in vitro assays provided evidence that EHD1 and SNX17 directly interact. We also found that inducing internalization of a SNX17 cargo receptor, low-density lipoprotein receptor-related protein 1 (LRP1), led to recruitment of cytoplasmic EHD1 to endosomal membranes. Moreover, surface rendering and quantification of overlap volumes indicated that SNX17 and EHD1 partially colocalize on endosomes and that this overlap further increases upon LRP1 internalization. Additionally, SNX17-containing endosomes were larger in EHD1-depleted cells than in WT cells, suggesting that EHD1 depletion impairs SNX17-mediated endosomal fission. Our findings help clarify our current understanding of endocytic trafficking, providing significant additional insight into the process of endosomal fission and connecting the sorting and fission machineries.
Collapse
Affiliation(s)
- Kanika Dhawan
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198 .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|
106
|
Li JG, Chiu J, Ramanjulu M, Blass BE, Praticò D. A pharmacological chaperone improves memory by reducing Aβ and tau neuropathology in a mouse model with plaques and tangles. Mol Neurodegener 2020; 15:1. [PMID: 31964406 PMCID: PMC6975032 DOI: 10.1186/s13024-019-0350-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 11/25/2022] Open
Abstract
Background The vacuolar protein sorting 35 (VPS35) is a major component of the retromer complex system, an ubiquitous multiprotein assembly responsible for sorting and trafficking protein cargos out of the endosomes. VPS35 can regulate APP metabolism and Aβ formation, and its levels are reduced in Alzheimer’s disease (AD) brains. We and others demonstrated that VPS35 genetic manipulation modulates the phenotype of mouse models of AD. However, the translational value of this observation remains to be investigated. Methods Triple transgenic mice were randomized to receive a pharmacological chaperone, which stabilizes the retromer complex, and the effect on their AD-like phenotype assessed. Results Compared with controls, treated mice had a significant improvement in learning and memory, an elevation of VPS35 levels, and improved synaptic integrity. Additionally, the same animals had a significant decrease in Aβ levels and deposition, reduced tau phosphorylation and less astrocytes activation. Conclusions Our study demonstrates that the enhancement of retromer function by pharmacological chaperones is a potentially novel and viable therapy against AD.
Collapse
Affiliation(s)
- Jian-Guo Li
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, MERB, suite 1160, Philadelphia, PA, 19140, USA
| | - Jin Chiu
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, MERB, suite 1160, Philadelphia, PA, 19140, USA
| | - Mercy Ramanjulu
- Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Benjamin E Blass
- Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, MERB, suite 1160, Philadelphia, PA, 19140, USA.
| |
Collapse
|
107
|
Tang FL, Zhao L, Zhao Y, Sun D, Zhu XJ, Mei L, Xiong WC. Coupling of terminal differentiation deficit with neurodegenerative pathology in Vps35-deficient pyramidal neurons. Cell Death Differ 2020; 27:2099-2116. [PMID: 31907392 PMCID: PMC7308361 DOI: 10.1038/s41418-019-0487-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Vps35 (vacuolar protein sorting 35) is a key component of retromer that regulates transmembrane protein trafficking. Dysfunctional Vps35 is a risk factor for neurodegenerative diseases, including Parkinson’s and Alzheimer’s diseases. Vps35 is highly expressed in developing pyramidal neurons, and its physiological role in developing neurons remains to be explored. Here, we provide evidence that Vps35 in embryonic neurons is necessary for axonal and dendritic terminal differentiation. Loss of Vps35 in embryonic neurons results in not only terminal differentiation deficits, but also neurodegenerative pathology, such as cortical brain atrophy and reactive glial responses. The atrophy of neocortex appears to be in association with increases in neuronal death, autophagosome proteins (LC3-II and P62), and neurodegeneration associated proteins (TDP43 and ubiquitin-conjugated proteins). Further studies reveal an increase of retromer cargo protein, sortilin1 (Sort1), in lysosomes of Vps35-KO neurons, and lysosomal dysfunction. Suppression of Sort1 diminishes Vps35-KO-induced dendritic defects. Expression of lysosomal Sort1 recapitulates Vps35-KO-induced phenotypes. Together, these results demonstrate embryonic neuronal Vps35’s function in terminal axonal and dendritic differentiation, reveal an association of terminal differentiation deficit with neurodegenerative pathology, and uncover an important lysosomal contribution to both events.
Collapse
Affiliation(s)
- Fu-Lei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia
| | - Lu Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.,Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Yang Zhao
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.,Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Dong Sun
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Xiao-Juan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia. .,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
108
|
Eleuteri S, Albanese A. VPS35-Based Approach: A Potential Innovative Treatment in Parkinson's Disease. Front Neurol 2019; 10:1272. [PMID: 31920908 PMCID: PMC6928206 DOI: 10.3389/fneur.2019.01272] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Several symptomatic treatments for Parkinson's disease (PD) are currently available. Still, the challenge today is to find a therapy that might reduce degeneration and slow down disease progression. The identification of pathogenic mutations in familial Parkinsonism (fPD) associated to the monogenic forms of PD provided pathophysiological insights and highlighted novel targets for therapeutic intervention. Mutations in the VPS35 gene have been associated with autosomal dominant fPD and a clinical phenotype indistinguishable from idiopathic PD. Although VPS35 mutations are relatively rare causes of PD, their study may help understanding specific cellular and molecular alterations that lead to accumulation α-synuclein in neurons of PD patients. Interacting with such mechanisms may provide innovative therapeutic approaches. We review here the evidence on the physiological role of VPS35 as a key intracellular trafficking protein controlling relevant neuronal functions. We further analyze VPS35 activity on α-synuclein degradation pathways that control the equilibrium between α-synuclein clearance and accumulation. Finally, we highlight the strategies for increasing VPS35 levels as a potential tool to treat PD.
Collapse
Affiliation(s)
- Simona Eleuteri
- Department of Neurology, Humanitas Research Hospital, Milan, Italy
| | - Alberto Albanese
- Department of Neurology, Humanitas Research Hospital, Milan, Italy.,Department of Neurology, Catholic University, Milan, Italy
| |
Collapse
|
109
|
Curnock R, Calcagni A, Ballabio A, Cullen PJ. TFEB controls retromer expression in response to nutrient availability. J Cell Biol 2019; 218:3954-3966. [PMID: 31694921 PMCID: PMC6891082 DOI: 10.1083/jcb.201903006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/05/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Endosomal recycling maintains the cell surface abundance of nutrient transporters for nutrient uptake, but how the cell integrates nutrient availability with recycling is less well understood. Here, in studying the recycling of human glutamine transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), SNAT1 (SLC38A1), and SNAT2 (SLC38A2), we establish that following amino acid restriction, the adaptive delivery of SNAT2 to the cell surface relies on retromer, a master conductor of endosomal recycling. Upon complete amino acid starvation or selective glutamine depletion, we establish that retromer expression is upregulated by transcription factor EB (TFEB) and other members of the MiTF/TFE family of transcription factors through association with CLEAR elements in the promoters of the retromer genes VPS35 and VPS26A TFEB regulation of retromer expression therefore supports adaptive nutrient acquisition through endosomal recycling.
Collapse
Affiliation(s)
- Rachel Curnock
- School of Biochemistry, University of Bristol, Bristol, UK
| | | | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, TX.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Peter J Cullen
- School of Biochemistry, University of Bristol, Bristol, UK
| |
Collapse
|
110
|
Pandey S, Dhusia K, Katara P, Singh S, Gautam B. An in silico analysis of deleterious single nucleotide polymorphisms and molecular dynamics simulation of disease linked mutations in genes responsible for neurodegenerative disorder. J Biomol Struct Dyn 2019; 38:4259-4272. [DOI: 10.1080/07391102.2019.1682047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sapna Pandey
- Department of Computational Biology & Bioinformatics, Jacob Institute of Biotechnology & Bio-Engineering, Sam Higginbottom University of Agriculture, Technology and Science (SHUATS), Allahabad, India
| | - Kalyani Dhusia
- Department of Computational Biology & Bioinformatics, Jacob Institute of Biotechnology & Bio-Engineering, Sam Higginbottom University of Agriculture, Technology and Science (SHUATS), Allahabad, India
- Department of Biomedical Engineering, Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Pramod Katara
- Centre of Bioinformatics, University of Allahabad, Allahabad, India
| | - Satendra Singh
- Department of Computational Biology & Bioinformatics, Jacob Institute of Biotechnology & Bio-Engineering, Sam Higginbottom University of Agriculture, Technology and Science (SHUATS), Allahabad, India
| | - Budhayash Gautam
- Department of Computational Biology & Bioinformatics, Jacob Institute of Biotechnology & Bio-Engineering, Sam Higginbottom University of Agriculture, Technology and Science (SHUATS), Allahabad, India
| |
Collapse
|
111
|
Ma M, Burd CG. Retrograde trafficking and plasma membrane recycling pathways of the budding yeast Saccharomyces cerevisiae. Traffic 2019; 21:45-59. [PMID: 31471931 DOI: 10.1111/tra.12693] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
The endosomal system functions as a network of protein and lipid sorting stations that receives molecules from endocytic and secretory pathways and directs them to the lysosome for degradation, or exports them from the endosome via retrograde trafficking or plasma membrane recycling pathways. Retrograde trafficking pathways describe endosome-to-Golgi transport while plasma membrane recycling pathways describe trafficking routes that return endocytosed molecules to the plasma membrane. These pathways are crucial for lysosome biogenesis, nutrient acquisition and homeostasis and for the physiological functions of many types of specialized cells. Retrograde and recycling sorting machineries of eukaryotic cells were identified chiefly through genetic screens using the budding yeast Saccharomyces cerevisiae system and discovered to be highly conserved in structures and functions. In this review, we discuss advances regarding retrograde trafficking and recycling pathways, including new discoveries that challenge existing ideas about the organization of the endosomal system, as well as how these pathways intersect with cellular homeostasis pathways.
Collapse
Affiliation(s)
- Mengxiao Ma
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut
| | - Christopher G Burd
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
112
|
Molecular identification of a BAR domain-containing coat complex for endosomal recycling of transmembrane proteins. Nat Cell Biol 2019; 21:1219-1233. [PMID: 31576058 PMCID: PMC6778059 DOI: 10.1038/s41556-019-0393-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
Protein trafficking requires coat complexes that couple recognition of sorting motifs in transmembrane cargos with biogenesis of transport carriers. The mechanisms of cargo transport through the endosomal network are poorly understood. Here, we identify a sorting motif for endosomal recycling of cargos including the cation-independent mannose-6-phosphate receptor and semaphorin 4C by the membrane tubulating BAR domain-containing sorting nexins SNX5 and SNX6. Crystal structures establish that this motif folds into a β-hairpin that binds a site in the SNX5/SNX6 phox homology domains. Over sixty cargos share this motif and require SNX5/SNX6 for their recycling. These include cargos involved in neuronal migration and a Drosophila snx6 mutant displays defects in axonal guidance. These studies identify a sorting motif and provide molecular insight into an evolutionary conserved coat complex, the ‘Endosomal SNX-BAR sorting complex for promoting exit 1’ (ESCPE-1), which couples sorting motif recognition to BAR domain-mediated biogenesis of cargo-enriched tubulo-vesicular transport carriers.
Collapse
|
113
|
Kvainickas A, Nägele H, Qi W, Dokládal L, Jimenez-Orgaz A, Stehl L, Gangurde D, Zhao Q, Hu Z, Dengjel J, De Virgilio C, Baumeister R, Steinberg F. Retromer and TBC1D5 maintain late endosomal RAB7 domains to enable amino acid-induced mTORC1 signaling. J Cell Biol 2019; 218:3019-3038. [PMID: 31431476 PMCID: PMC6719456 DOI: 10.1083/jcb.201812110] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/30/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
Retromer is an evolutionarily conserved multiprotein complex that orchestrates the endocytic recycling of integral membrane proteins. Here, we demonstrate that retromer is also required to maintain lysosomal amino acid signaling through mTORC1 across species. Without retromer, amino acids no longer stimulate mTORC1 translocation to the lysosomal membrane, which leads to a loss of mTORC1 activity and increased induction of autophagy. Mechanistically, we show that its effect on mTORC1 activity is not linked to retromer's role in the recycling of transmembrane proteins. Instead, retromer cooperates with the RAB7-GAP TBC1D5 to restrict late endosomal RAB7 into microdomains that are spatially separated from the amino acid-sensing domains. Upon loss of retromer, RAB7 expands into the ragulator-decorated amino acid-sensing domains and interferes with RAG-GTPase and mTORC1 recruitment. Depletion of retromer in Caenorhabditis elegans reduces mTORC1 signaling and extends the lifespan of the worms, confirming an evolutionarily conserved and unexpected role for retromer in the regulation of mTORC1 activity and longevity.
Collapse
Affiliation(s)
- Arunas Kvainickas
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Heike Nägele
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Wenjing Qi
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ladislav Dokládal
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Ana Jimenez-Orgaz
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Luca Stehl
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Dipak Gangurde
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Qian Zhao
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Zehan Hu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Ralf Baumeister
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Florian Steinberg
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| |
Collapse
|
114
|
Abstract
Protein coats are supramolecular complexes that assemble on the cytosolic face of membranes to promote cargo sorting and transport carrier formation in the endomembrane system of eukaryotic cells. Several types of protein coats have been described, including COPI, COPII, AP-1, AP-2, AP-3, AP-4, AP-5, and retromer, which operate at different stages of the endomembrane system. Defects in these coats impair specific transport pathways, compromising the function and viability of the cells. In humans, mutations in subunits of these coats cause various congenital diseases that are collectively referred to as coatopathies. In this article, we review the fundamental properties of protein coats and the diseases that result from mutation of their constituent subunits.
Collapse
Affiliation(s)
- Esteban C Dell'Angelica
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
115
|
Chen K, Healy MD, Collins BM. Towards a molecular understanding of endosomal trafficking by Retromer and Retriever. Traffic 2019; 20:465-478. [DOI: 10.1111/tra.12649] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Kai‐En Chen
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| | - Michael D. Healy
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| | - Brett M. Collins
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
116
|
Vagnozzi AN, Praticò D. Endosomal sorting and trafficking, the retromer complex and neurodegeneration. Mol Psychiatry 2019; 24:857-868. [PMID: 30120416 PMCID: PMC6378136 DOI: 10.1038/s41380-018-0221-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 02/08/2023]
Abstract
The retromer is a highly conserved multimeric protein complex present in all eukaryotic cells whose activity is essential for regulating the recycling and retrieval of numerous protein cargos from the endosome to trans-Golgi network or the cell surface. In recent years, molecular and genomic studies have provided evidence that aberrant regulation of endosomal protein sorting and trafficking secondary to a dysfunction of the retromer complex could be implicated in the pathogenesis of several neurodegenerative diseases. Thus, deficiency or mutations in one or more protein components of the retromer leads to increased accumulation of protein aggregates, as well as enhanced cellular neurotoxicity. In this review, we will discuss the structure and function of the retromer complex and its neurobiology, its relevance to key molecules involved in neurodegeneration and the potential role that it plays in the development of two major neurodegenerative disorders, Parkinson's disease and Alzheimer's disease. Finally, we will discuss the viability of targeting the retromer via pharmacological chaperones or genetic approaches to enhance or restore its function as a novel and unifying disease-modifying strategy against these diseases.
Collapse
Affiliation(s)
- Alana N. Vagnozzi
- Alzheimer’s Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 191040
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 191040, USA.
| |
Collapse
|
117
|
Finetti F, Cassioli C, Cianfanelli V, Onnis A, Paccagnini E, Kabanova A, Baldari CT. The intraflagellar transport protein IFT20 controls lysosome biogenesis by regulating the post-Golgi transport of acid hydrolases. Cell Death Differ 2019; 27:310-328. [PMID: 31142807 DOI: 10.1038/s41418-019-0357-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/16/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023] Open
Abstract
The assembly and function of the primary cilium depends on multimolecular intraflagellar transport (IFT) complexes that shuttle their cargo along the axonemal microtubules through their interaction with molecular motors. The IFT system has been moreover recently implicated in a reciprocal interplay between autophagy and ciliogenesis. We have previously reported that IFT20 and other components of the IFT complexes participate in the assembly of the immune synapse in the non-ciliated T cell, suggesting that other cellular processes regulated by the IFT system in ciliated cells, including autophagy, may be shared by cells lacking a cilium. Starting from the observation of a defect in autophagic clearance and an accumulation of lipid droplets in IFT20-deficient T cells, we show that IFT20 is required for lysosome biogenesis and function by controlling the lysosomal targeting of acid hydrolases. This function involves its ability to regulate the retrograde traffic of the cation-independent mannose-6-phosphate receptor (CI-MPR) to the trans-Golgi network, which is achieved by coupling recycling CI-MPRs to the microtubule motor dynein. Consistent with the lysosomal defect, an upregulation of the TFEB-dependent expression of the lysosomal gene network can be observed in IFT20-deficient cells, which is associated with defective tonic T-cell antigen receptor signaling and mTOR activity. We additionally show that the lysosome-related function of IFT20 extends to non-ciliated cells other than T cells, as well as to ciliated cells. Our findings provide the first evidence that a component of the IFT system that controls ciliogenesis is implicated in the biogenesis of lysosomes.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Chiara Cassioli
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Valentina Cianfanelli
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Anna Onnis
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Eugenio Paccagnini
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Anna Kabanova
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| |
Collapse
|
118
|
Vorberg IM. All the Same? The Secret Life of Prion Strains within Their Target Cells. Viruses 2019; 11:v11040334. [PMID: 30970585 DOI: 10.3390/v11040334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 01/23/2023] Open
Abstract
Prions are infectious β-sheet-rich protein aggregates composed of misfolded prion protein (PrPSc) that do not possess coding nucleic acid. Prions replicate by recruiting and converting normal cellular PrPC into infectious isoforms. In the same host species, prion strains target distinct brain regions and cause different disease phenotypes. Prion strains are associated with biophysically distinct PrPSc conformers, suggesting that strain properties are enciphered within alternative PrPSc quaternary structures. So far it is unknown how prion strains target specific cells and initiate productive infections. Deeper mechanistic insight into the prion life cycle came from cell lines permissive to a range of different prion strains. Still, it is unknown why certain cell lines are refractory to infection by one strain but permissive to another. While pharmacologic and genetic manipulations revealed subcellular compartments involved in prion replication, little is known about strain-specific requirements for endocytic trafficking pathways. This review summarizes our knowledge on how prions replicate within their target cells and on strain-specific differences in prion cell biology.
Collapse
Affiliation(s)
- Ina M Vorberg
- German Center for Neurodegenerative Diseases (DZNE e.V.), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
- Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany.
| |
Collapse
|
119
|
USP32 regulates late endosomal transport and recycling through deubiquitylation of Rab7. Nat Commun 2019; 10:1454. [PMID: 30926795 PMCID: PMC6440979 DOI: 10.1038/s41467-019-09437-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/06/2019] [Indexed: 12/26/2022] Open
Abstract
The endosomal system is a highly dynamic multifunctional organelle, whose complexity is regulated in part by reversible ubiquitylation. Despite the wide-ranging influence of ubiquitin in endosomal processes, relatively few enzymes utilizing ubiquitin have been described to control endosome integrity and function. Here we reveal the deubiquitylating enzyme (DUB) ubiquitin-specific protease 32 (USP32) as a powerful player in this context. Loss of USP32 inhibits late endosome (LE) transport and recycling of LE cargos, resulting in dispersion and swelling of the late compartment. Using SILAC-based ubiquitome profiling we identify the small GTPase Rab7—the logistical centerpiece of LE biology—as a substrate of USP32. Mechanistic studies reveal that LE transport effector RILP prefers ubiquitylation-deficient Rab7, while retromer-mediated LE recycling benefits from an intact cycle of Rab7 ubiquitylation. Collectively, our observations suggest that reversible ubiquitylation helps switch Rab7 between its various functions, thereby maintaining global spatiotemporal order in the endosomal system. Though ubiquitin is known to broadly influence endosomal trafficking, few ubiquitin-utilizing enzymes targeting endosomal regulators are known. Here, the authors find that the deubiquitylating enzyme (DUB) USP32 influences endosomal membrane dynamics by deubiquitinating Rab7.
Collapse
|
120
|
Farmer T, O'Neill KL, Naslavsky N, Luo X, Caplan S. Retromer facilitates the localization of Bcl-xL to the mitochondrial outer membrane. Mol Biol Cell 2019; 30:1138-1146. [PMID: 30840537 PMCID: PMC6724524 DOI: 10.1091/mbc.e19-01-0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The anti-apoptotic Bcl-2 family protein Bcl-xL plays a critical role in cell survival by protecting the integrity of the mitochondrial outer membrane (MOM). The mechanism through which Bcl-xL is recruited to the MOM has not been fully discerned. The retromer is a conserved endosomal scaffold complex involved in membrane trafficking. Here we identify VPS35 and VPS26, two core components of the retromer, as novel regulators of Bcl-xL. We observed interactions and colocalization between Bcl-xL, VPS35, VPS26, and MICAL-L1, a protein involved in recycling endosome biogenesis that also interacts with the retromer. We also found that upon VPS35 depletion, levels of nonmitochondrial Bcl-xL were increased. In addition, retromer-depleted cells displayed more rapid Bax activation and apoptosis. These results suggest that the retromer regulates apoptosis by facilitating Bcl-xL's transport to the MOM. Importantly, our studies suggest a previously uncharacterized relationship between the machineries of cell death/survival and endosomal trafficking.
Collapse
Affiliation(s)
- Trey Farmer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Katelyn L O'Neill
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Xu Luo
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870.,Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| |
Collapse
|
121
|
Cui Y, Yang Z, Teasdale RD. A role of GCC88 in the retrograde transport of CI-M6PR and the maintenance of lysosomal activity. Cell Biol Int 2019; 43:1234-1244. [PMID: 30791178 DOI: 10.1002/cbin.11118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/17/2019] [Indexed: 11/10/2022]
Abstract
GCC88 is a golgin coiled-coil protein at the trans-Golgi (TGN) that functions as a tethering factor for the endosome-derived retrograde transport vesicles. Here, we demonstrate that GCC88 is required for the endosome-to-TGN retrograde transport of the cation-independent mannose 6-phosphate receptor (CI-M6PR). The knockout of GCC88 perturbs the retrieval of CI-M6PR and decreases its cellular level at the steady state, which causes the improper processing of newly synthesized cathepsin-D, a lysosomal hydrolase dependent on CI-M6PR for its delivery to lysosomes. At the whole cell level, the knockout of GCC88 reduces the lysosomal proteolytic capacity but does not impair of the efficiency of autophagy within these cells.
Collapse
Affiliation(s)
- Yi Cui
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhe Yang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rohan D Teasdale
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
122
|
Seaman MNJ. Back From the Brink: Retrieval of Membrane Proteins From Terminal Compartments: Unexpected Pathways for Membrane Protein Retrieval From Vacuoles and Endolysosomes. Bioessays 2019; 41:e1800146. [PMID: 30706963 DOI: 10.1002/bies.201800146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/03/2018] [Indexed: 11/12/2022]
Abstract
It has long been believed that membrane proteins present in degradative compartments such as endolysosomes or vacuoles would be destined for destruction. Now however, it appears that mechanisms and machinery exist in simple eukaryotes such as yeast and more complex organisms such as mammals that can rescue potentially "doomed" membrane proteins by retrieving them from these "late" compartments and recycling them back to the Golgi complex. In yeast, a sorting nexin dimer containing Snx4p can recognize and retrieve the Atg27p membrane protein while in mammals, the AP5 complex (with SPG11 and SPG15) directs the recycling of Golgi-localized proteins along with the cation-independent mannose 6-phosphate receptor (CIMPR). Although the respective machinery is different, there is much commonality between yeast and mammals regarding the mechanisms of retrieval and the physiological importance of these late recycling pathways.
Collapse
Affiliation(s)
- Matthew N J Seaman
- University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrookes Hospital, CB2 0XY, UK
| |
Collapse
|
123
|
Makhoul C, Gosavi P, Duffield R, Delbridge B, Williamson NA, Gleeson PA. Intersectin-1 interacts with the golgin GCC88 to couple the actin network and Golgi architecture. Mol Biol Cell 2019; 30:370-386. [PMID: 30540523 PMCID: PMC6589577 DOI: 10.1091/mbc.e18-05-0313] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/19/2018] [Accepted: 12/04/2018] [Indexed: 12/19/2022] Open
Abstract
The maintenance of the Golgi ribbon relies on a dynamic balance between the actin and microtubule networks; however, the pathways controlling actin networks remain poorly defined. Previously, we showed that the trans-Golgi network (TGN) membrane tether/golgin, GCC88, modulates the Golgi ribbon architecture. Here, we show that dispersal of the Golgi ribbon by GCC88 is dependent on actin and the involvement of nonmuscle myosin IIA. We have identified the long isoform of intersectin-1 (ITSN-1), a guanine nucleotide exchange factor for Cdc42, as a novel Golgi component and an interaction partner of GCC88 responsible for mediating the actin-dependent dispersal of the Golgi ribbon. We show that perturbation of Golgi morphology by changes in membrane flux, mediated by silencing the retromer subunit Vps26, or in a model of neurodegeneration, induced by Tau overexpression, are also dependent on the ITSN-1-GCC88 interaction. Overall, our study reveals a role for a TGN golgin and ITSN-1 in linking to the actin cytoskeleton and regulating the balance between a compact Golgi ribbon and a dispersed Golgi, a pathway with relevance to pathophysiological conditions.
Collapse
Affiliation(s)
- Christian Makhoul
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Prajakta Gosavi
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Regina Duffield
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Bronwen Delbridge
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Nicholas A. Williamson
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A. Gleeson
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
124
|
Rahman AA, Morrison BE. Contributions of VPS35 Mutations to Parkinson's Disease. Neuroscience 2019; 401:1-10. [PMID: 30660673 DOI: 10.1016/j.neuroscience.2019.01.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/18/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's Disease (PD) is a multi-system neurodegenerative disease where approximately 90% of cases are idiopathic. The remaining 10% of the cases can be traced to a genetic origin and research has largely focused on these associated genes to gain a better understanding of the molecular and cellular pathogenesis for PD. The gene encoding vacuolar protein sorting protein 35 (VPS35) has been definitively linked to late onset familial PD following the identification of a point mutation (D620N) as the causal agent in a Swiss family. Since its discovery, numerous studies have been undertaken to characterize the role of VPS35 in cellular processes and efforts have been directed toward understanding the perturbations caused by the D620N mutation. In this review, we examine what is currently known about VPS35, which has pleiotropic effects, as well as proposed mechanisms of pathogenesis by the D620N mutation. A brief survey of other VPS35 polymorphisms is also provided. Lastly, model systems that are being utilized for these investigations and possible directions for future research are discussed.
Collapse
Affiliation(s)
- Abir A Rahman
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA; Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, USA
| | - Brad E Morrison
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA; Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|
125
|
Rudenok MM, Alieva AK, Nikolaev MA, Kolacheva AA, Ugryumov MV, Pchelina SN, Slominsky PA, Shadrina MI. Possible Involvement of Genes Related to Lysosomal Storage Disorders in the Pathogenesis of Parkinson’s Disease. Mol Biol 2019. [DOI: 10.1134/s002689331901014x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
126
|
Cui Y, Carosi JM, Yang Z, Ariotti N, Kerr MC, Parton RG, Sargeant TJ, Teasdale RD. Retromer has a selective function in cargo sorting via endosome transport carriers. J Cell Biol 2018; 218:615-631. [PMID: 30559172 PMCID: PMC6363445 DOI: 10.1083/jcb.201806153] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/22/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
The molecular actions of retromer in the endolysosomal system remain unclear and controversial. Cui et al. demonstrate the essential role of retromer in the selective incorporation of cargo into a specific type of endosome transport carrier and the maintenance of lysosomal function. Retromer is a peripheral membrane protein complex that coordinates multiple vesicular trafficking events within the endolysosomal system. Here, we demonstrate that retromer is required for the maintenance of normal lysosomal morphology and function. The knockout of retromer subunit Vps35 causes an ultrastructural alteration in lysosomal structure and aberrant lysosome function, leading to impaired autophagy. At the whole-cell level, knockout of retromer Vps35 subunit reduces lysosomal proteolytic capacity as a consequence of the improper processing of lysosomal hydrolases, which is dependent on the trafficking of the cation-independent mannose 6-phosphate receptor (CI-M6PR). Incorporation of CI-M6PR into endosome transport carriers via a retromer-dependent process is restricted to those tethered by GCC88 but not golgin-97 or golgin-245. Finally, we show that this retromer-dependent retrograde cargo trafficking pathway requires SNX3, but not other retromer-associated cargo binding proteins, such as SNX27 or SNX-BAR proteins. Therefore, retromer does contribute to the retrograde trafficking of CI-M6PR required for maturation of lysosomal hydrolases and lysosomal function.
Collapse
Affiliation(s)
- Yi Cui
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Julian M Carosi
- Hopwood Centre for Neurobiology, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Zhe Yang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Ariotti
- Institute for Molecular Biosciences and Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Markus C Kerr
- Institute for Molecular Biosciences and Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Robert G Parton
- Institute for Molecular Biosciences and Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Timothy J Sargeant
- Hopwood Centre for Neurobiology, Nutrition and Metabolism Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Rohan D Teasdale
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
127
|
Cullen PJ, Steinberg F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat Rev Mol Cell Biol 2018; 19:679-696. [PMID: 30194414 DOI: 10.1038/s41580-018-0053-7] [Citation(s) in RCA: 388] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Newly endocytosed integral cell surface proteins are typically either directed for degradation or subjected to recycling back to the plasma membrane. The sorting of integral cell surface proteins, including signalling receptors, nutrient transporters, ion channels, adhesion molecules and polarity markers, within the endolysosomal network for recycling is increasingly recognized as an essential feature in regulating the complexities of physiology at the cell, tissue and organism levels. Historically, endocytic recycling has been regarded as a relatively passive process, where the majority of internalized integral proteins are recycled via a nonspecific sequence-independent 'bulk membrane flow' pathway. Recent work has increasingly challenged this view. The discovery of sequence-specific sorting motifs and the identification of cargo adaptors and associated coat complexes have begun to uncover the highly orchestrated nature of endosomal cargo recycling, thereby providing new insight into the function and (patho)physiology of this process.
Collapse
Affiliation(s)
- Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK.
| | - Florian Steinberg
- Center for Biological Systems Analysis, Albert Ludwigs Universitaet Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
128
|
Cai X, Dong J, Liu J, Zheng H, Kaweeteerawat C, Wang F, Ji Z, Li R. Multi-hierarchical profiling the structure-activity relationships of engineered nanomaterials at nano-bio interfaces. Nat Commun 2018; 9:4416. [PMID: 30356046 PMCID: PMC6200803 DOI: 10.1038/s41467-018-06869-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022] Open
Abstract
Increasing concerns over the possible risks of nanotechnology necessitates breakthroughs in structure-activity relationship (SAR) analyses of engineered nanomaterials (ENMs) at nano-bio interfaces. However, current nano-SARs are often based on univariate assessments and fail to provide tiered views on ENM-induced bio-effects. Here we report a multi-hierarchical nano-SAR assessment for a representative ENM, Fe2O3, by metabolomics and proteomics analyses. The established nano-SAR profile allows the visualizing of the contributions of seven basic properties of Fe2O3 to its diverse bio-effects. For instance, although surface reactivity is responsible for Fe2O3-induced cell migration, the inflammatory effects of Fe2O3 are determined by aspect ratio (nanorods) or surface reactivity (nanoplates). These nano-SARs are examined in THP-1 cells and animal lungs, which allow us to decipher the detailed mechanisms including NLRP3 inflammasome pathway and monocyte chemoattractant protein-1-dependent signaling. This study provides more insights for nano-SARs, and may facilitate the tailored design of ENMs to render them desired bio-effects.
Collapse
Affiliation(s)
- Xiaoming Cai
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| | - Jun Dong
- Wuhan Academy of Agricultural Science, Wuhan, Hubei 430000 China
| | - Jing Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, 116023 China
| | - Huizhen Zheng
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| | - Chitrada Kaweeteerawat
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Klong Nueng, 12120 Thailand
| | - Fangjun Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, 116023 China
| | - Zhaoxia Ji
- California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
- Living Proof, Inc., Cambridge, MA 02142 United States
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, School of Public Health, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123 China
| |
Collapse
|
129
|
Stroupe C. This Is the End: Regulation of Rab7 Nucleotide Binding in Endolysosomal Trafficking and Autophagy. Front Cell Dev Biol 2018; 6:129. [PMID: 30333976 PMCID: PMC6176412 DOI: 10.3389/fcell.2018.00129] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/14/2018] [Indexed: 01/07/2023] Open
Abstract
Rab7 – or in yeast, Ypt7p – governs membrane trafficking in the late endocytic and autophagic pathways. Rab7 also regulates mitochondrion-lysosome contacts, the sites of mitochondrial fission. Like all Rab GTPases, Rab7 cycles between an “active” GTP-bound form that binds downstream effectors – e.g., the HOPS and retromer complexes and the dynactin-binding Rab-interacting lysosomal protein (RILP) – and an “inactive” GDP-bound form that cannot bind effectors. Accessory proteins regulate the nucleotide binding state of Rab7: guanine nucleotide exchange factors (GEFs) stimulate exchange of bound GDP for GTP, resulting in Rab7 activation, whereas GTPase activating proteins (GAPs) boost Rab7’s GTP hydrolysis activity, thereby inactivating Rab7. This review will discuss the GEF and GAPs that control Rab7 nucleotide binding, and thus regulate Rab7’s activity in endolysosomal trafficking and autophagy. It will also consider how bacterial pathogens manipulate Rab7 nucleotide binding to support intracellular invasion and immune evasion.
Collapse
Affiliation(s)
- Christopher Stroupe
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
130
|
Kimura N, Yanagisawa K. Traffic jam hypothesis: Relationship between endocytic dysfunction and Alzheimer's disease. Neurochem Int 2018; 119:35-41. [DOI: 10.1016/j.neuint.2017.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/30/2017] [Accepted: 07/07/2017] [Indexed: 01/07/2023]
|
131
|
Reduced retromer function results in the accumulation of amyloid-beta oligomers. Mol Cell Neurosci 2018; 93:18-26. [PMID: 30257187 DOI: 10.1016/j.mcn.2018.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 11/24/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of multiple cognitive functions. Accumulation of amyloid beta oligomers (oAβ) play a major role in the neurotoxicity associated with the disease process. One of the early affected brain regions is the hippocampus, wherein a reduction of the vacuolar protein sorting-associated protein 35 (VPS35), the core protein comprising the retromer complex involved in cellular cargo sorting, has been identified. To investigate the role of the retromer function on the accumulation and clearance of oAβ, we reduced retromer function by selectively inhibiting VPS35 gene expression using siRNA in differentiated neuronal SH-SY5Y cells. As cell-to-cell transfer of oAβ to new brain regions is believed to be important for disease progression we investigated the effect of VPS35 reduction both in cells with direct uptake of oAβ and in cells receiving oAβ from donor cells. We demonstrate that reduced retromer function increases oAβ accumulation in both cell systems, both the number of cells containing intracellular oAβ and the amount within them. This effect was shown at different time points and regardless if the oAβ originated from the extracellular milieu or via a direct neuronal cell-to-cell transfer. Interestingly, not only did reduced VPS35 cause oAβ accumulation, but oAβ treatment alone also lead to a reduction of VPS35 protein content. The accumulated oAβ seems to co-localize with VPS35 and early endosome markers. Together, these findings provide evidence that reduced retromer function decreases the ability for neurons to transport and clear neurotoxic oAβ received through different routes resulting in the accumulation of oAβ. Thus, enhancing retromer function may be a potential therapeutic strategy to slow down the pathophysiology associated with the progression of AD.
Collapse
|
132
|
Hoyer MJ, Chitwood PJ, Ebmeier CC, Striepen JF, Qi RZ, Old WM, Voeltz GK. A Novel Class of ER Membrane Proteins Regulates ER-Associated Endosome Fission. Cell 2018; 175:254-265.e14. [PMID: 30220460 PMCID: PMC6195207 DOI: 10.1016/j.cell.2018.08.030] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 05/01/2018] [Accepted: 08/15/2018] [Indexed: 11/24/2022]
Abstract
Endoplasmic reticulum (ER) membrane contact sites (MCSs) mark positions where endosomes undergo fission for cargo sorting. To define the role of ER at this unique MCS, we targeted a promiscuous biotin ligase to cargo-sorting domains on endosome buds. This strategy identified the ER membrane protein TMCC1, a member of a conserved protein family. TMCC1 concentrates at the ER-endosome MCSs that are spatially and temporally linked to endosome fission. When TMCC1 is depleted, endosome morphology is normal, buds still form, but ER-associated bud fission and subsequent cargo sorting to the Golgi are impaired. We find that the endosome-localized actin regulator Coronin 1C is required for ER-associated fission of actin-dependent cargo-sorting domains. Coronin 1C is recruited to endosome buds independently of TMCC1, while TMCC1/ER recruitment requires Coronin 1C. This link between TMCC1 and Coronin 1C suggests that the timing of TMCC1-dependent ER recruitment is tightly regulated to occur after cargo has been properly sequestered into the bud.
Collapse
Affiliation(s)
- Melissa J Hoyer
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA
| | - Patrick J Chitwood
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA
| | - Christopher C Ebmeier
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA
| | - Jonathan F Striepen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA
| | - Robert Z Qi
- Division of Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - William M Old
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA
| | - Gia K Voeltz
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, University of Colorado-Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
133
|
Gireud-Goss M, Reyes S, Wilson M, Farley M, Memarzadeh K, Srinivasan S, Sirisaengtaksin N, Yamashita S, Tsunoda S, Lang FF, Waxham MN, Bean AJ. Distinct mechanisms enable inward or outward budding from late endosomes/multivesicular bodies. Exp Cell Res 2018; 372:1-15. [PMID: 30144444 DOI: 10.1016/j.yexcr.2018.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
Regulating the residence time of membrane proteins on the cell surface can modify their response to extracellular cues and allow for cellular adaptation in response to changing environmental conditions. The fate of membrane proteins that are internalized from the plasma membrane and arrive at the limiting membrane of the late endosome/multivesicular body (MVB) is dictated by whether they remain on the limiting membrane, bud into internal MVB vesicles, or bud outwardly from the membrane. The molecular details underlying the disposition of membrane proteins that transit this pathway and the mechanisms regulating these trafficking events are unclear. We established a cell-free system that reconstitutes budding of membrane protein cargo into internal MVB vesicles and onto vesicles that bud outwardly from the MVB membrane. Both budding reactions are cytosol-dependent and supported by Saccharomyces cerevisiae (yeast) cytosol. We observed that inward and outward budding from the MVB membrane are mechanistically distinct but may be linked, such that inhibition of inward budding triggers a re-routing of cargo from inward to outward budding vesicles, without affecting the number of vesicles that bud outwardly from MVBs.
Collapse
Affiliation(s)
- Monica Gireud-Goss
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Sahily Reyes
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Marenda Wilson
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Madeline Farley
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Kimiya Memarzadeh
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | | | - Natalie Sirisaengtaksin
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| | - Shinji Yamashita
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - M Neal Waxham
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Andrew J Bean
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Biochemistry and Cell Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA; Department of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
134
|
Cataldi S, Follett J, Fox JD, Tatarnikov I, Kadgien C, Gustavsson EK, Khinda J, Milnerwood AJ, Farrer MJ. Altered dopamine release and monoamine transporters in Vps35 p.D620N knock-in mice. NPJ PARKINSONS DISEASE 2018; 4:27. [PMID: 30155515 PMCID: PMC6104078 DOI: 10.1038/s41531-018-0063-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 11/09/2022]
Abstract
Vacuolar protein sorting 35 (VPS35) is a core component of the retromer trimer required for endosomal membrane-associated protein trafficking. The discovery of a missense mutation, Vps35 p.D620N implicates retromer dysfunction in the pathogenesis of Parkinson's disease (PD). We have characterized a knock-in mouse with a Vps35 p.D620N substitution (hereafter referred to as VKI) at 3 months of age. Standardized behavioral testing did not observe overt movement disorder. Tyrosine hydroxylase (TH)-positive nigral neuron counts and terminal expression in striata were comparable across genotypes. Fast scan cyclic voltammetry revealed increased dopamine release in VKI striatal slices. While extracellular dopamine collected via striatal microdialysis of freely moving animals was comparable across genotypes, the ratio of dopamine metabolites to dopamine suggests increased dopamine turnover in VKI homozygous mice. Western blot of striatal proteins revealed a genotype-dependent decrease in dopamine transporter (DAT) along with an increase in vesicular monoamine transporter 2 (VMAT2), albeit independent of changes in other synaptic markers. The reduction in DAT was further supported by immunohistochemical analysis. The data show that the dopaminergic system of VKI mice is profoundly altered relative to wild-type littermates. We conclude early synaptic dysfunction contributes to age-related pathophysiology in the nigrostriatal system that may lead to parkinsonism in man.
Collapse
Affiliation(s)
- Stefano Cataldi
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Jordan Follett
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Jesse D Fox
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Igor Tatarnikov
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Chelsie Kadgien
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Emil K Gustavsson
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada.,2Department of Neurology, St. Olav's Hospital, Trondheim, Norway
| | - Jaskaran Khinda
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Austen J Milnerwood
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| | - Matthew J Farrer
- 1Centre for Applied Neurogenetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
135
|
Singh V, Yang J, Yin J, Cole R, Tse M, Berman DE, Small SA, Petsko G, Donowitz M. Cholera toxin inhibits SNX27-retromer-mediated delivery of cargo proteins to the plasma membrane. J Cell Sci 2018; 131:jcs218610. [PMID: 30030371 PMCID: PMC6127729 DOI: 10.1242/jcs.218610] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/22/2018] [Indexed: 12/14/2022] Open
Abstract
Cholera toxin (CT) causes severe diarrhea by increasing intracellular cAMP leading to a PKA-dependent increase in Cl- secretion through CFTR and decreased Na+ absorption through inhibition of Na+/H+ exchanger 3 (NHE3; also known as SLC9A3). The mechanism(s) by which CT inhibits NHE3 is partially understood, although no drug therapy has been successful at reversing this inhibition. We now describe that CT phosphorylates an amino acid in the PDZ domain of SNX27, which inhibits SNX27-mediated trafficking of NHE3 from the early endosomes to the plasma membrane (PM), and contributes to reduced basal NHE3 activity through a mechanism that involves reduced PM expression and reduced endocytic recycling. Importantly, mutagenesis studies (Ser to Asp) showed that the effect of this phosphorylation of SNX27 phenocopies the effects seen upon loss of SNX27 function, affecting PM trafficking of cargo proteins that bind SNX27-retromer. Additionally, CT destabilizes retromer function by decreasing the amount of core retromer proteins. These effects of CT can be partially rescued by enhancing retromer stability by using 'pharmacological chaperones'. Moreover, pharmacological chaperones can be used to increase basal and cholera toxin-inhibited NHE3 activity and fluid absorption by intestinal epithelial cells.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Varsha Singh
- Departments of Medicine and Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jianbo Yang
- Departments of Medicine and Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jianyi Yin
- Departments of Medicine and Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert Cole
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ming Tse
- Departments of Medicine and Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Diego E Berman
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Pathology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Scott A Small
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Pathology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Gregory Petsko
- Helen and Robert Appel Alzheimer's Disease Research Institute and Department of Neurology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Mark Donowitz
- Departments of Medicine and Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
136
|
Audano M, Schneider A, Mitro N. Mitochondria, lysosomes, and dysfunction: their meaning in neurodegeneration. J Neurochem 2018; 147:291-309. [DOI: 10.1111/jnc.14471] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Matteo Audano
- DiSFeB; Dipartimento di Scienze Farmacologiche e Biomolecolari; Università degli Studi di Milano; Milano Italy
| | - Anja Schneider
- German Center for Neurodegenerative Diseases; DZNE; Bonn Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry; University Clinic; Bonn Germany
| | - Nico Mitro
- DiSFeB; Dipartimento di Scienze Farmacologiche e Biomolecolari; Università degli Studi di Milano; Milano Italy
| |
Collapse
|
137
|
Wang J, Fedoseienko A, Chen B, Burstein E, Jia D, Billadeau DD. Endosomal receptor trafficking: Retromer and beyond. Traffic 2018; 19:578-590. [PMID: 29667289 PMCID: PMC6043395 DOI: 10.1111/tra.12574] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/17/2022]
Abstract
The tubular endolysosomal network is a quality control system that ensures the proper delivery of internalized receptors to specific subcellular destinations in order to maintain cellular homeostasis. Although retromer was originally described in yeast as a regulator of endosome-to-Golgi receptor recycling, mammalian retromer has emerged as a central player in endosome-to-plasma membrane recycling of a variety of receptors. Over the past decade, information regarding the mechanism by which retromer facilitates receptor trafficking has emerged, as has the identification of numerous retromer-associated molecules including the WASH complex, sorting nexins (SNXs) and TBC1d5. Moreover, the recent demonstration that several SNXs can directly interact with retromer cargo to facilitate endosome-to-Golgi retrieval has provided new insight into how these receptors are trafficked in cells. The mechanism by which SNX17 cargoes are recycled out of the endosomal system was demonstrated to involve a retromer-like complex termed the retriever, which is recruited to WASH positive endosomes through an interaction with the COMMD/CCDC22/CCDC93 (CCC) complex. Lastly, the mechanisms by which bacterial and viral pathogens highjack this complex sorting machinery in order to escape the endolysosomal system or remain hidden within the cells are beginning to emerge. In this review, we will highlight recent studies that have begun to unravel the intricacies by which the retromer and associated molecules contribute to receptor trafficking and how deregulation at this sorting domain can contribute to disease or facilitate pathogen infection.
Collapse
Affiliation(s)
- Jing Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, Division of Neurology, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Alina Fedoseienko
- Division of Oncology Research, Department of Biochemistry and Molecular Biology, and Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Bayou Chen
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa 50011, USA
| | - Ezra Burstein
- Department of Internal Medicine, and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, Division of Neurology, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Daniel D. Billadeau
- Division of Oncology Research, Department of Biochemistry and Molecular Biology, and Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
138
|
Farmer T, Naslavsky N, Caplan S. Tying trafficking to fusion and fission at the mighty mitochondria. Traffic 2018; 19:569-577. [PMID: 29663589 PMCID: PMC6043374 DOI: 10.1111/tra.12573] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/11/2018] [Accepted: 04/11/2018] [Indexed: 01/03/2023]
Abstract
The mitochondrion is a unique organelle that serves as the main site of ATP generation needed for energy in the cell. However, mitochondria also play essential roles in cell death through apoptosis and necrosis, as well as a variety of crucial functions related to stress regulation, autophagy, lipid synthesis and calcium storage. There is a growing appreciation that mitochondrial function is regulated by the dynamics of its membrane fusion and fission; longer, fused mitochondria are optimal for ATP generation, whereas fission of mitochondria facilitates mitophagy and cell division. Despite the significance of mitochondrial homeostasis for such crucial cellular events, the intricate regulation of mitochondrial fusion and fission is only partially understood. Until very recently, only a single mitochondrial fission protein had been identified. Moreover, only now have researchers turned to address the upstream machinery that regulates mitochondrial fusion and fission proteins. Herein, we review the known GTPases involved in mitochondrial fusion and fission, but also highlight recent studies that address the mechanisms by which these GTPases are regulated. In particular, we draw attention to a substantial new body of literature linking endocytic regulatory proteins, such as the retromer VPS35 cargo selection complex subunit, to mitochondrial homeostasis. These recent studies suggest that relationships and cross-regulation between endocytic and mitochondrial pathways may be more widespread than previously assumed.
Collapse
Affiliation(s)
- Trey Farmer
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, Nebraska
| | - Naava Naslavsky
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, Nebraska
| | - Steve Caplan
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, Nebraska
- The Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
139
|
Endosomal Retrieval of Cargo: Retromer Is Not Alone. Trends Cell Biol 2018; 28:807-822. [PMID: 30072228 DOI: 10.1016/j.tcb.2018.06.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/15/2018] [Accepted: 06/22/2018] [Indexed: 11/20/2022]
Abstract
Endosomes are major protein sorting stations in cells. Endosomally localised multi-protein complexes sort integral proteins, including signaling receptors, nutrient transporters, adhesion molecules, and lysosomal hydrolase receptors, for lysosomal degradation or conversely for retrieval and subsequent recycling to various membrane compartments. Correct endosomal sorting of these proteins is essential for maintaining cellular homeostasis, with defects in endosomal sorting implicated in various human pathologies including neurodegenerative disorders. Retromer, an ancient multi-protein complex, is essential for the retrieval and recycling of hundreds of transmembrane proteins. While retromer is a major player in endosomal retrieval and recycling, several studies have recently identified retrieval mechanisms that are independent of retromer. Here, we review endosomal retrieval complexes, with a focus on recently discovered retromer-independent mechanisms.
Collapse
|
140
|
Chang F, Li N, Yan K, Huang Y, Xu H, Liu Y. Luminal/extracellular domains of chimeric CI-M6PR-C proteins interfere with their retrograde endosome-to-TGN trafficking in the transient expression system. J Biomed Res 2018; 32:245-256. [PMID: 29988026 PMCID: PMC6117607 DOI: 10.7555/jbr.32.20180044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The membrane trafficking of cation-independent mannose 6-phosphate receptor (CI-M6PR) between the trans-Golgi network (TGN) and endosomal compartments is not only critical for maintaining lysosomal function but also a well-known event for understanding molecular and cellular mechanisms in retrograde endosome-to-TGN trafficking. Although it has been well established in literature that the C-terminus of bovine CI-M6PR determines its retrograde trafficking, it remains unclear whether the luminal domain of the protein plays a role on these sorting events. In this study, we found that partial deletion of luminal domain of human CI-M6PR mistargeted the mutant protein to non-TGN compartments. Moreover, replacing the luminal domain of both bovine and human CI-M6PR with that from irrelevant membrane proteins such as CD8 or Tac also altered the TGN targeting of the chimeric proteins. On the other hand, only short sequence from HA fused with the transmembrane domain and C-terminus of the receptor, HA-hCI-M6PR-tail, resulted in its preferential targeting to TGN as for the full length receptor, strongly suggesting that sorting of the receptor may be influenced by luminal sequence. Furthermore, using this luminal truncated form of HA-hCI-M6PR as a model cargo, we found that the trafficking of the chimeric protein was regulated by the retromer complex through interacting with SNX5. In conclusion, our study strongly suggested that the disrupted luminal domain from hCI-M6PR or other irrelevant membrane proteins interfere with the process of membrane trafficking and TGN targeting of CI-M6PR.
Collapse
Affiliation(s)
- Fei Chang
- . Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Na Li
- . Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Kang Yan
- . Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yumin Huang
- . Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hongfei Xu
- . Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yongjian Liu
- . Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
,. Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
,Corresponding author: Dr. Yongjian Liu, Jiangsu Key Laboratory of Xenotransplanation, Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA, Tel: +86-25-86869442,
| |
Collapse
|
141
|
Naslavsky N, Caplan S. The enigmatic endosome - sorting the ins and outs of endocytic trafficking. J Cell Sci 2018; 131:131/13/jcs216499. [PMID: 29980602 DOI: 10.1242/jcs.216499] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The early endosome (EE), also known as the sorting endosome (SE) is a crucial station for the sorting of cargoes, such as receptors and lipids, through the endocytic pathways. The term endosome relates to the receptacle-like nature of this organelle, to which endocytosed cargoes are funneled upon internalization from the plasma membrane. Having been delivered by the fusion of internalized vesicles with the EE or SE, cargo molecules are then sorted to a variety of endocytic pathways, including the endo-lysosomal pathway for degradation, direct or rapid recycling to the plasma membrane, and to a slower recycling pathway that involves a specialized form of endosome known as a recycling endosome (RE), often localized to the perinuclear endocytic recycling compartment (ERC). It is striking that 'the endosome', which plays such essential cellular roles, has managed to avoid a precise description, and its characteristics remain ambiguous and heterogeneous. Moreover, despite the rapid advances in scientific methodologies, including breakthroughs in light microscopy, overall, the endosome remains poorly defined. This Review will attempt to collate key characteristics of the different types of endosomes and provide a platform for discussion of this unique and fascinating collection of organelles. Moreover, under-developed, poorly understood and important open questions will be discussed.
Collapse
Affiliation(s)
- Naava Naslavsky
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Steve Caplan
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA .,The Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
142
|
A versatile nanobody-based toolkit to analyze retrograde transport from the cell surface. Proc Natl Acad Sci U S A 2018; 115:E6227-E6236. [PMID: 29915061 DOI: 10.1073/pnas.1801865115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Retrograde transport of membranes and proteins from the cell surface to the Golgi and beyond is essential to maintain homeostasis, compartment identity, and physiological functions. To study retrograde traffic biochemically, by live-cell imaging or by electron microscopy, we engineered functionalized anti-GFP nanobodies (camelid VHH antibody domains) to be bacterially expressed and purified. Tyrosine sulfation consensus sequences were fused to the nanobody for biochemical detection of trans-Golgi arrival, fluorophores for fluorescence microscopy and live imaging, and APEX2 (ascorbate peroxidase 2) for electron microscopy and compartment ablation. These functionalized nanobodies are specifically captured by GFP-modified reporter proteins at the cell surface and transported piggyback to the reporters' homing compartments. As an application of this tool, we have used it to determine the contribution of adaptor protein-1/clathrin in retrograde transport kinetics of the mannose-6-phosphate receptors from endosomes back to the trans-Golgi network. Our experiments establish functionalized nanobodies as a powerful tool to demonstrate and quantify retrograde transport pathways.
Collapse
|
143
|
Tábara LC, Vicente JJ, Biazik J, Eskelinen EL, Vincent O, Escalante R. Vacuole membrane protein 1 marks endoplasmic reticulum subdomains enriched in phospholipid synthesizing enzymes and is required for phosphoinositide distribution. Traffic 2018; 19:624-638. [DOI: 10.1111/tra.12581] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Luis-Carlos Tábara
- Instituto de Investigaciones Biomédicas Alberto Sols, C.S.I.C./U.A.M.; Madrid Spain
| | - Juan-Jesús Vicente
- Department of Physiology and Biophysics, School of Medicine; University of Washington; Seattle Washington
| | - Joanna Biazik
- Department of Biosciences; University of Helsinki; Helsinki Finland
| | | | - Olivier Vincent
- Instituto de Investigaciones Biomédicas Alberto Sols, C.S.I.C./U.A.M.; Madrid Spain
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas Alberto Sols, C.S.I.C./U.A.M.; Madrid Spain
| |
Collapse
|
144
|
MicroRNA 199a-5p Attenuates Retrograde Transport and Protects against Toxin-Induced Inhibition of Protein Biosynthesis. Mol Cell Biol 2018; 38:MCB.00548-17. [PMID: 29555727 DOI: 10.1128/mcb.00548-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 03/08/2018] [Indexed: 01/09/2023] Open
Abstract
Retrograde transport (RT) allows cells to retrieve receptors and other cellular cargoes for delivery to the Golgi apparatus, contributing to the maintenance of cellular homeostasis. This transport route is also commonly used by several bacterial toxins to exert their deleterious actions on eukaryotic cells. While the retrograde transport process has been well characterized, the contribution of microRNAs (miRNAs) in regulating this cellular transport mechanism remains unknown. Here, we determined that mir-199a and mir-199b, members of the intronic miRNA family, coordinate genes regulating RT and endosome trafficking. We demonstrate that miR-199a-5p attenuates the expression of Vps26A, Rab9B, and M6PR, thereby controlling RT from endosomes to the trans-Golgi network (TGN). Importantly, we found that overexpression of a Vps26A construct resistant to the inhibitory action of miR-199a-5p abrogates the effect of miR-199a-5p on RT. Finally, we demonstrate that miR-199-5p overexpression attenuates Shiga toxin type 1 (Stx1)-mediated inhibition of protein biosynthesis. In summary, our work identifies the first noncoding RNA that influences RT and reduces the inhibition of protein biosynthesis caused by bacterial toxins.
Collapse
|
145
|
Bingol B. Autophagy and lysosomal pathways in nervous system disorders. Mol Cell Neurosci 2018; 91:167-208. [PMID: 29729319 DOI: 10.1016/j.mcn.2018.04.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionarily conserved pathway for delivering cytoplasmic cargo to lysosomes for degradation. In its classically studied form, autophagy is a stress response induced by starvation to recycle building blocks for essential cellular processes. In addition, autophagy maintains basal cellular homeostasis by degrading endogenous substrates such as cytoplasmic proteins, protein aggregates, damaged organelles, as well as exogenous substrates such as bacteria and viruses. Given their important role in homeostasis, autophagy and lysosomal machinery are genetically linked to multiple human disorders such as chronic inflammatory diseases, cardiomyopathies, cancer, and neurodegenerative diseases. Multiple targets within the autophagy and lysosomal pathways offer therapeutic opportunities to benefit patients with these disorders. Here, I will summarize the mechanisms of autophagy pathways, the evidence supporting a pathogenic role for disturbed autophagy and lysosomal degradation in nervous system disorders, and the therapeutic potential of autophagy modulators in the clinic.
Collapse
Affiliation(s)
- Baris Bingol
- Genentech, Inc., Department of Neuroscience, 1 DNA Way, South San Francisco 94080, United States.
| |
Collapse
|
146
|
Elwell C, Engel J. Emerging Role of Retromer in Modulating Pathogen Growth. Trends Microbiol 2018; 26:769-780. [PMID: 29703496 DOI: 10.1016/j.tim.2018.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/21/2018] [Accepted: 04/02/2018] [Indexed: 12/20/2022]
Abstract
Intracellular pathogens have developed elegant mechanisms to modulate host endosomal trafficking. The highly conserved retromer pathway has emerged as an important target of viruses and intravacuolar bacteria. Some pathogens require retromer function to survive. For others, retromer activity restricts intracellular growth; these pathogens must disrupt retromer function to survive. In this review, we discuss recent paradigm changes to the current model for retromer assembly and cargo selection. We highlight how the study of pathogen effectors has contributed to these fundamental insights, with a special focus on the biology and structure of two recently described bacterial effectors, Chlamydia trachomatis IncE and Legionella pneumophila RidL. These two pathogens employ distinct strategies to target retromer components and overcome restriction of intracellular growth imposed by retromer.
Collapse
Affiliation(s)
- Cherilyn Elwell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joanne Engel
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
147
|
Tammineni P, Jeong YY, Feng T, Aikal D, Cai Q. Impaired axonal retrograde trafficking of the retromer complex augments lysosomal deficits in Alzheimer's disease neurons. Hum Mol Genet 2018; 26:4352-4366. [PMID: 28973312 DOI: 10.1093/hmg/ddx321] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/09/2017] [Indexed: 01/13/2023] Open
Abstract
Lysosomal proteolysis is essential for the quality control of intracellular components and the maintenance of cellular homeostasis. Lysosomal alterations have been implicated as one of the main cellular defects contributing to the onset and progression of Alzheimer's disease (AD). However, the mechanism underlying lysosomal deficits in AD remains poorly understood. Here, we reveal that lysosomal deficits are attributed to retromer dysfunction induced by altered retromer trafficking in the axon of AD-related mutant human amyloid precursor protein (hAPP) transgenic (Tg) mouse neurons. We demonstrate that retrograde transport of retromer is impaired, leading to its significant reduction in the soma and abnormal retention within late endosomes in distal axons of mutant hAPP neurons. Therefore, retromer-mediated endosome-to-Golgi retrieval of cation-independent mannose-6-phosphate receptors (CI-MPR) in the soma is disrupted in mutant hAPP neurons, causing defects in lysosome biogenesis. Such defects result in protease deficiency in lysosomes and impaired lysosomal proteolysis, as evidenced by aberrant accumulation of sequestered substrates within lysosomes. Intriguingly, enhancement of retrograde transport in mutant hAPP neurons facilitates the trafficking of axonal retromer toward the soma and thus enhances protease transport to lysosomes, thereby restoring lysosomal proteolytic activity. Taken together, our study provides new insights into the regulation of retromer trafficking through retrograde axonal transport to fulfil its function in promoting lysosome biogenesis in the soma, suggesting a potential approach for rescuing lysosomal proteolysis deficits in AD.
Collapse
Affiliation(s)
- Prasad Tammineni
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yu Young Jeong
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Tuancheng Feng
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Daniyal Aikal
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
148
|
Human Papillomavirus 16 Infection Induces VAP-Dependent Endosomal Tubulation. J Virol 2018; 92:JVI.01514-17. [PMID: 29321327 DOI: 10.1128/jvi.01514-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/20/2017] [Indexed: 02/02/2023] Open
Abstract
Human papillomavirus (HPV) infection involves complex interactions with the endocytic transport machinery, which ultimately facilitates the entry of the incoming viral genomes into the trans-Golgi network (TGN) and their subsequent nuclear entry during mitosis. The endosomal pathway is a highly dynamic intracellular transport system, which consists of vesicular compartments and tubular extensions, although it is currently unclear whether incoming viruses specifically alter the endocytic machinery. In this study, using MICAL-L1 as a marker for tubulating endosomes, we show that incoming HPV-16 virions induce a profound alteration in global levels of endocytic tubulation. In addition, we also show a critical requirement for the endoplasmic reticulum (ER)-anchored protein VAP in this process. VAP plays an essential role in actin nucleation and endosome-to-Golgi transport. Indeed, the loss of VAP results in a dramatic decrease in the level of endosomal tubulation induced by incoming HPV-16 virions. This is also accompanied by a marked reduction in virus infectivity. In VAP knockdown cells, we see that the defect in virus trafficking occurs after capsid disassembly but prior to localization at the trans-Golgi network, with the incoming virion-transduced DNA accumulating in Vps29/TGN46-positive hybrid vesicles. Taken together, these studies demonstrate that infection with HPV-16 virions induces marked alterations of endocytic transport pathways, some of which are VAP dependent and required for the endosome-to-Golgi transport of the incoming viral L2/DNA complex.IMPORTANCE Human papillomavirus infectious entry involves multiple interactions with the endocytic transport machinery. In this study, we show that incoming HPV-16 virions induce a dramatic increase in endocytic tubulation. This tubulation requires ER-associated VAP, which plays a critical role in ensuring the delivery of cargoes from the endocytic compartments to the trans-Golgi network. Indeed, the loss of VAP blocks HPV infectious entry at a step after capsid uncoating but prior to localization at the trans-Golgi network. These results define a critical role for ER-associated VAP in endocytic tubulation and in HPV-16 infectious entry.
Collapse
|
149
|
VPS35 depletion does not impair presynaptic structure and function. Sci Rep 2018; 8:2996. [PMID: 29445238 PMCID: PMC5812998 DOI: 10.1038/s41598-018-20448-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/18/2018] [Indexed: 12/03/2022] Open
Abstract
The endosomal system is proposed as a mediator of synaptic vesicle recycling, but the molecular recycling mechanism remains largely unknown. Retromer is a key protein complex which mediates endosomal recycling in eukaryotic cells, including neurons. Retromer is important for brain function and mutations in retromer genes are linked to neurodegenerative diseases. In this study, we aimed to determine the role of retromer in presynaptic structure and function. We assessed the role of retromer by knocking down VPS35, the core subunit of retromer, in primary hippocampal mouse neurons. VPS35 depletion led to retromer dysfunction, measured as a decrease in GluA1 at the plasma membrane, and bypassed morphological defects previously described in chronic retromer depletion models. We found that retromer is localized at the mammalian presynaptic terminal. However, VPS35 depletion did not alter the presynaptic ultrastructure, synaptic vesicle release or retrieval. Hence, we conclude that retromer is present in the presynaptic terminal but it is not essential for the synaptic vesicle cycle. Nonetheless, the presynaptic localization of VPS35 suggests that retromer-dependent endosome sorting could take place for other presynaptic cargo.
Collapse
|
150
|
Carpier JM, Zucchetti AE, Bataille L, Dogniaux S, Shafaq-Zadah M, Bardin S, Lucchino M, Maurin M, Joannas LD, Magalhaes JG, Johannes L, Galli T, Goud B, Hivroz C. Rab6-dependent retrograde traffic of LAT controls immune synapse formation and T cell activation. J Exp Med 2018; 215:1245-1265. [PMID: 29440364 PMCID: PMC5881459 DOI: 10.1084/jem.20162042] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 11/30/2017] [Accepted: 01/11/2018] [Indexed: 12/24/2022] Open
Abstract
The adapter molecule linker for activation of T cells (LAT) orchestrates the formation of signalosomes upon T cell receptor (TCR) stimulation. LAT is present in different intracellular pools and is dynamically recruited to the immune synapse upon stimulation. However, the intracellular traffic of LAT and its function in T lymphocyte activation are ill defined. We show herein that LAT, once internalized, transits through the Golgi-trans-Golgi network (TGN), where it is repolarized to the immune synapse. This retrograde transport of LAT depends on the small GTPase Rab6 and the target soluble N-ethylmaleimide-sensitive factor attachment protein receptor (t-SNARE) Syntaxin-16, two regulators of the endosome-to-Golgi/TGN retrograde transport. We also show in vitro in Syntaxin-16- or Rab6-silenced human cells and in vivo in CD4+ T lymphocytes of the Rab6 knockout mouse that this retrograde traffic controls TCR stimulation. These results establish that the retrograde traffic of LAT from the plasma membrane to the Golgi-TGN controls the polarized delivery of LAT at the immune synapse and T lymphocyte activation.
Collapse
Affiliation(s)
- Jean-Marie Carpier
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Andres E Zucchetti
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Laurence Bataille
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Stéphanie Dogniaux
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Massiullah Shafaq-Zadah
- Cellular and Chemical Biology of Membranes and Therapeutic Delivery Unit, Institut Curie, Paris Sciences and Lettres Research University, INSERM U1143, CNRS UMR 3666, Paris, France
| | - Sabine Bardin
- Molecular Mechanisms of Intracellular Transport Group, Institut Curie, Paris Sciences and Lettres Research University, CNRS UMR 144, Paris, France
| | - Marco Lucchino
- Cellular and Chemical Biology of Membranes and Therapeutic Delivery Unit, Institut Curie, Paris Sciences and Lettres Research University, INSERM U1143, CNRS UMR 3666, Paris, France
| | - Mathieu Maurin
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Leonel D Joannas
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Joao Gamelas Magalhaes
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| | - Ludger Johannes
- Cellular and Chemical Biology of Membranes and Therapeutic Delivery Unit, Institut Curie, Paris Sciences and Lettres Research University, INSERM U1143, CNRS UMR 3666, Paris, France
| | - Thierry Galli
- Center of Psychiatry and Neurosciences, Membrane Traffic in Health and Diseased Brain, Université Paris Descartes, Sorbonne Paris Cité, INSERM ERL U950, Paris, France
| | - Bruno Goud
- Molecular Mechanisms of Intracellular Transport Group, Institut Curie, Paris Sciences and Lettres Research University, CNRS UMR 144, Paris, France
| | - Claire Hivroz
- Crosstalk between T Cells and Dendritic Cells Group, Institut Curie, Paris Sciences and Lettres Research University, INSERM U932, Paris, France
| |
Collapse
|