1
|
Wang X, Li T, Guo Y, Chen XW. License to drive: Receptor-mediated ER exit of proteins and lipids. Curr Opin Cell Biol 2025; 94:102501. [PMID: 40117676 DOI: 10.1016/j.ceb.2025.102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
The secretory pathway, which begins at the endoplasmic reticulum (ER) through the COPII complex, is responsible for transporting proteins and lipid carriers to various destined cellular compartments or extracellular space. The fundamental mechanism by which the COPII operates is evolutionarily conserved. Nevertheless, the vast diversity of mammalian cargos poses significant challenges to the secretory pathway, especially considering the intricate physiology in vivo. Particularly, certain physiologically essential cargos, including procollagen and lipoproteins, appear to be oversized for these canonical carriers, implying the need for additional sophisticated regulation at the onset step so-called ER exit. Emerging evidence highlights the critical role of cargo receptors in selective sorting for ER export, illuminating the complex biology of the trafficking dynamics, which holds broad implications for human health and diseases.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Tiantian Li
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yusong Guo
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Shenzhen Research Institute, Hong Kong University of Science and Technology, Shenzhen, 518057, China; Thrust of Bioscience and Biomedical Engineering, Hong Kong University of Science and Technology, Guangzhou, 511453, China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing, 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China; Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
2
|
Bögershausen N, Cavdarli B, Nagai TH, Milev MP, Wolff A, Mehranfar M, Schmidt J, Choudhary D, Gutiérrez-Gutiérrez Ó, Cyganek L, Saint-Dic D, Zibat A, Köhrer K, Wollenweber TE, Wieczorek D, Altmüller J, Borodina T, Kaçar D, Haliloğlu G, Li Y, Thiel C, Sacher M, Knapik EW, Yigit G, Wollnik B. SEC24C deficiency causes trafficking and glycosylation abnormalities in an epileptic encephalopathy with cataracts and dyserythropoeisis. JCI Insight 2025; 10:e173484. [PMID: 40131364 DOI: 10.1172/jci.insight.173484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
As a major component of intracellular trafficking, the coat protein complex II (COPII) is indispensable for cellular function during embryonic development and throughout life. The 4 SEC24 proteins (A-D) are essential COPII components involved in cargo selection and packaging. A human disorder corresponding to alterations of SEC24 function is currently known only for SEC24D. Here, we reported that biallelic loss of SEC24C leads to a syndrome characterized by primary microcephaly, brain anomalies, epilepsy, hearing loss, liver dysfunction, anemia, and cataracts in an extended consanguineous family with 4 affected individuals. We showed that knockout of sec24C in zebrafish recapitulated important aspects of the human phenotype. SEC24C-deficient fibroblasts displayed alterations in the expression of several COPII components as well as impaired anterograde trafficking to the Golgi, indicating a severe impact on COPII function. Transcriptome analysis revealed that SEC24C deficiency also affected the proteasome and autophagy pathways. Moreover, a shift in the N-glycosylation pattern and deregulation of the N-glycosylation pathway suggested a possible secondary alteration of protein glycosylation, linking the described disorder with the congenital disorders of glycosylation.
Collapse
Affiliation(s)
- Nina Bögershausen
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Büsranur Cavdarli
- Department of Medical Genetics, Ankara Bilkent City Hospital, Ankara, Turkey
- Department of Medical Genetics, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Taylor H Nagai
- Vanderbilt Genetics Institute and Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Miroslav P Milev
- Department of Biology, Concordia University, Montréal, Québec, Canada
| | - Alexander Wolff
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Göttingen, Göttingen, Germany
| | - Mahsa Mehranfar
- Department of Chemistry and Biochemistry, Concordia University, Montréal, Québec, Canada
| | - Julia Schmidt
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Dharmendra Choudhary
- Vanderbilt Genetics Institute and Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Óscar Gutiérrez-Gutiérrez
- Stem Cell Unit, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neuroinflammation and Automated Microscopy TNM, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neuroinflammation and Automated Microscopy TNM, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Lower Saxony, Germany
- Cluster of Excellence Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany
| | - Djenann Saint-Dic
- Department of Biology, Concordia University, Montréal, Québec, Canada
| | - Arne Zibat
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Karl Köhrer
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Center (BMFZ), and
| | - Tassilo E Wollenweber
- Genomics & Transcriptomics Laboratory, Biological and Medical Research Center (BMFZ), and
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Janine Altmüller
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Tatiana Borodina
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dilek Kaçar
- Department of Pediatric Hematology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Göknur Haliloğlu
- Division of Pediatric Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yun Li
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Christian Thiel
- Center for Pediatric and Adolescent Medicine, Department I, Heidelberg University, Heidelberg, Germany
| | - Michael Sacher
- Department of Biology, Concordia University, Montréal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Ela W Knapik
- Vanderbilt Genetics Institute and Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Lower Saxony, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Lower Saxony, Lower Saxony, Germany
- Cluster of Excellence Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
3
|
Ke L, Liu J, Feng G, Li X, Zhang Y, Zhang S, Ma X, Di Q. Effects of acute PM 2.5 purification on cognitive function and underlying mechanisms: Evidence from integrating alternative splicing into multi-omics. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137214. [PMID: 39823879 DOI: 10.1016/j.jhazmat.2025.137214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
The relationship between fine particulate matter (PM2.5) and cognition has been extensively investigated. However, the causal impact of acute PM2.5 purification on cognition improvement and the underlying biological mechanisms remain relatively opaque. Our double-blinded randomized controlled trial assessed the impact of acute PM2.5 purification on executive function, underpinned by multi-omics approaches including alternative splicing (AS) analysis. A total of 93 participants experienced a two-hour exposure to either reduced and normal PM2.5 levels. We measured the cognition of healthy young adults, collected peripheral blood before and after intervention, and performed multi-omics analysis including transcriptomics, metabolomics, and proteomics. Results indicated that reducing PM2.5 by 1 μg/m3 was associated with a 0.10 % (95 % CI: [0.18 %, 0.01 %]; p = 0.031) improvement in executive function. Notably, we identified 96 AS events without concurrent transcriptional amount alterations. Multi-layered omics analyses revealed disrupted pathways in hypoxia, mitochondrial function and energy metabolism, and immune responses, validated by ELISA and biochemical assay. These findings demonstrated short-term improvements of cognition following PM2.5 purification and provide mechanistic understandings of PM2.5-induced cognition alterations. This study underscores the significance of incorporating AS in the molecular framework of multi-omics research by exploring variable exon splicing, which could enrich multi-omics analysis methodologies and expose to broader audience.
Collapse
Affiliation(s)
- Limei Ke
- School of Medicine, Tsinghua University, Beijing 100084, China; School of Biomedical Engineering, Tsinghua University, Beijing 100084, China.
| | - Jianxiu Liu
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China.
| | - Guoqing Feng
- School of Medicine, Tsinghua University, Beijing 100084, China; School of Biomedical Engineering, Tsinghua University, Beijing 100084, China.
| | - Xingtian Li
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China.
| | - Yao Zhang
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China; Soochow College, Soochow University, Suzhou 215006, China.
| | - Shiqi Zhang
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China.
| | - Xindong Ma
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China; Institute for Healthy China, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
4
|
Ronzier E, Satpute-Krishnan P. TMED9 coordinates the clearance of misfolded GPI-anchored proteins out of the ER and into the Golgi. PLoS Biol 2025; 23:e3003084. [PMID: 40203033 PMCID: PMC12052135 DOI: 10.1371/journal.pbio.3003084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/05/2025] [Accepted: 02/25/2025] [Indexed: 04/11/2025] Open
Abstract
The p24-family member, TMED9, has recently emerged as a player in secretory pathway protein quality control (PQC) that influences the trafficking and degradation of misfolded proteins. Here, we show that TMED9 plays a central role in the PQC of GPI-anchored proteins (GPI-APs). Typically, upon release from the endoplasmic reticulum (ER)-resident chaperone calnexin, misfolded GPI-APs traffic to the Golgi by an ER-export pathway called Rapid ER stress-induced Export (RESET). From the Golgi, they access the plasma membrane where they are rapidly internalized for lysosomal degradation. We used biochemical and imaging approaches in cultured cells to demonstrate that at steady-state, the majority of misfolded GPI-APs reside in the ER in association with calnexin and TMED9. During RESET, they dissociate from calnexin and increase their association with TMED9. Inhibition of TMED9's function through siRNA-induced depletion or chemical inhibitor, BRD4780, blocked ER-export of misfolded GPI-APs. In contrast, TMED9-inhibition did not prevent ER-export of wild-type GPI-APs, indicating a specific role for TMED9 in GPI-AP PQC. Intriguingly, we discovered that acute treatment with BRD4780 induced a shift in TMED9 localization away from the ER to the downstream Golgi cisternae and blocked the RESET pathway. Upon removal of BRD4780 following acute treatment, TMED9 regained access to the ER where TMED9 was able to associate with the RESET substrate and restore the RESET pathway. These results suggest that TMED9 plays a requisite role in RESET by capturing misfolded GPI-APs that are released by calnexin within the ER and conveying them to the Golgi.
Collapse
Affiliation(s)
- Elsa Ronzier
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Prasanna Satpute-Krishnan
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
5
|
Downes KW, Zanetti G. Mechanisms of COPII coat assembly and cargo recognition in the secretory pathway. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00839-y. [PMID: 40133632 PMCID: PMC7617623 DOI: 10.1038/s41580-025-00839-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
One third of all proteins in eukaryotes transit between the endoplasmic reticulum (ER) and the Golgi to reach their functional destination inside or outside of the cell. During export, secretory proteins concentrate at transitional zones of the ER known as ER exit sites, where they are packaged into transport carriers formed by the highly conserved coat protein complex II (COPII). Despite long-standing knowledge of many of the fundamental pathways that govern traffic in the early secretory pathway, we still lack a complete mechanistic model to explain how the various steps of COPII-mediated ER exit are regulated to efficiently transport diverse cargoes. In this Review, we discuss the current understanding of the mechanisms underlying COPII-mediated vesicular transport, highlighting outstanding knowledge gaps. We focus on how coat assembly and disassembly dictate carrier morphogenesis, how COPII selectively recruits a vast number of cargo and cargo adaptors, and finally discuss how COPII mechanisms in mammals might have adapted to enable transport of large proteins.
Collapse
Affiliation(s)
- Katie W Downes
- Institute of Structural and Molecular Biology, UCL, London, UK
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK
- The Francis Crick Institute, London, UK
| | - Giulia Zanetti
- Institute of Structural and Molecular Biology, UCL, London, UK.
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
6
|
Ronzier E, Satpute-Krishnan P. TMED9 coordinates the clearance of misfolded GPI-anchored proteins out of the endoplasmic reticulum and into the Golgi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.27.615420. [PMID: 39974996 PMCID: PMC11838446 DOI: 10.1101/2024.09.27.615420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The p24-family member, TMED9, has recently emerged as a player in secretory pathway protein quality control (PQC) that influences the trafficking and degradation of misfolded proteins. Here we show that TMED9 plays a central role in the PQC of GPI-anchored proteins (GPI-APs). Typically, upon release from the endoplasmic reticulum (ER)-resident chaperone calnexin, misfolded GPI-APs traffic to the Golgi by an ER-export pathway called Rapid ER stress-induced Export (RESET). From the Golgi, they access the plasma membrane where they are rapidly internalized for lysosomal degradation. We used biochemical and imaging approaches in cultured cells to demonstrate that at steady-state, the majority of misfolded GPI-APs reside in the ER in association with calnexin and TMED9. During RESET, they dissociate from calnexin and increase their association with TMED9. Inhibition of TMED9's function through siRNA-induced depletion or chemical inhibitor, BRD4780, blocked ER-export of misfolded GPI-APs. By contrast, TMED9-inhibition did not prevent ER-export of wild type GPI-APs, indicating a specific role for TMED9 in GPI-AP PQC. Intriguingly, we discovered that acute treatment with BRD4780 induced a shift in TMED9 localization away from the ER to the downstream Golgi cisternae and blocked the RESET pathway. Upon removal of BRD4780 following acute treatment, TMED9 regained access to the ER where TMED9 was able to associate with the RESET substrate and restore the RESET pathway. These results suggest that TMED9 plays a requisite role in RESET by capturing misfolded GPI-APs that are released by calnexin within the ER and conveying them to the Golgi.
Collapse
|
7
|
Prokisch S, Büttner S. Partitioning into ER membrane microdomains impacts autophagic protein turnover during cellular aging. Sci Rep 2024; 14:13653. [PMID: 38871812 DOI: 10.1038/s41598-024-64493-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
Eukaryotic membranes are compartmentalized into distinct micro- and nanodomains that rearrange dynamically in response to external and internal cues. This lateral heterogeneity of the lipid bilayer and associated clustering of distinct membrane proteins contribute to the spatial organization of numerous cellular processes. Here, we show that membrane microdomains within the endoplasmic reticulum (ER) of yeast cells are reorganized during metabolic reprogramming and aging. Using biosensors with varying transmembrane domain length to map lipid bilayer thickness, we demonstrate that in young cells, microdomains of increased thickness mainly exist within the nuclear ER, while progressing cellular age drives the formation of numerous microdomains specifically in the cortical ER. Partitioning of biosensors with long transmembrane domains into these microdomains increased protein stability and prevented autophagic removal. In contrast, reporters with short transmembrane domains progressively accumulated at the membrane contact site between the nuclear ER and the vacuole, the so-called nucleus-vacuole junction (NVJ), and were subjected to turnover via selective microautophagy occurring specifically at these sites. Reporters with long transmembrane domains were excluded from the NVJ. Our data reveal age-dependent rearrangement of the lateral organization of the ER and establish transmembrane domain length as a determinant of membrane contact site localization and autophagic degradation.
Collapse
Affiliation(s)
- Simon Prokisch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden
| | - Sabrina Büttner
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691, Stockholm, Sweden.
| |
Collapse
|
8
|
Yang K, Feng Z, Pastor-Pareja JC. p24-Tango1 interactions ensure ER-Golgi interface stability and efficient transport. J Cell Biol 2024; 223:e202309045. [PMID: 38470362 PMCID: PMC10932740 DOI: 10.1083/jcb.202309045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/07/2024] [Accepted: 02/05/2024] [Indexed: 03/13/2024] Open
Abstract
The eukaryotic p24 family, consisting of α-, β-, γ- and δ-p24 subfamilies, has long been known to be involved in regulating secretion. Despite increasing interest in these proteins, fundamental questions remain about their role. Here, we systematically investigated Drosophila p24 proteins. We discovered that members of all four p24 subfamilies are required for general secretion and that their localizations between ER exit site (ERES) and Golgi are interdependent in an α→βδ→γ sequence. We also found that localization of p24 proteins and ERES determinant Tango1 requires interaction through their respective GOLD and SH3 lumenal domains, with Tango1 loss sending p24 proteins to the plasma membrane and vice versa. Finally, we show that p24 loss expands the COPII zone at ERES and increases the number of ER-Golgi vesicles, supporting a restrictive role of p24 proteins on vesicle budding for efficient transport. Our results reveal Tango1-p24 interplay as central to the generation of a stable ER-Golgi interface.
Collapse
Affiliation(s)
- Ke Yang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhi Feng
- School of Life Sciences, Tsinghua University, Beijing, China
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Institute of Neurosciences, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
9
|
Knopf JD, Steigleder SS, Korn F, Kühnle N, Badenes M, Tauber M, Theobald SJ, Rybniker J, Adrain C, Lemberg MK. RHBDL4-triggered downregulation of COPII adaptor protein TMED7 suppresses TLR4-mediated inflammatory signaling. Nat Commun 2024; 15:1528. [PMID: 38453906 PMCID: PMC10920636 DOI: 10.1038/s41467-024-45615-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
The toll-like receptor 4 (TLR4) is a central regulator of innate immunity that primarily recognizes bacterial lipopolysaccharide cell wall constituents to trigger cytokine secretion. We identify the intramembrane protease RHBDL4 as a negative regulator of TLR4 signaling. We show that RHBDL4 triggers degradation of TLR4's trafficking factor TMED7. This counteracts TLR4 transport to the cell surface. Notably, TLR4 activation mediates transcriptional upregulation of RHBDL4 thereby inducing a negative feedback loop to reduce TLR4 trafficking to the plasma membrane. This secretory cargo tuning mechanism prevents the over-activation of TLR4-dependent signaling in an in vitro Mycobacterium tuberculosis macrophage infection model and consequently alleviates septic shock in a mouse model. A hypomorphic RHBDL4 mutation linked to Kawasaki syndrome, an ill-defined inflammatory disorder in children, further supports the pathophysiological relevance of our findings. In this work, we identify an RHBDL4-mediated axis that acts as a rheostat to prevent over-activation of the TLR4 pathway.
Collapse
Affiliation(s)
- Julia D Knopf
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Susanne S Steigleder
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Friederike Korn
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Nathalie Kühnle
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Marina Badenes
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
- Faculty of Veterinary Medicine, Lusofona University and Faculty of Veterinary Nursing, Polytechnic Institute of Lusofonia, Lisbon, Portugal
| | - Marina Tauber
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931, Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931, Cologne, Germany
| | - Colin Adrain
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Marius K Lemberg
- Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany.
- Center for Biochemistry and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Roberts BS, Mitra D, Abishek S, Beher R, Satpute-Krishnan P. The p24-family and COPII subunit SEC24C facilitate the clearance of alpha1-antitrypsin Z from the endoplasmic reticulum to lysosomes. Mol Biol Cell 2024; 35:ar45. [PMID: 38294851 PMCID: PMC10916869 DOI: 10.1091/mbc.e23-06-0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/01/2024] Open
Abstract
A subpopulation of the alpha-1-antitrypsin misfolding Z mutant (ATZ) is cleared from the endoplasmic reticulum (ER) via an ER-to-lysosome-associated degradation (ERLAD) pathway. Here, we report that the COPII subunit SEC24C and the p24-family of proteins facilitate the clearance of ATZ via ERLAD. In addition to the previously reported ERLAD components calnexin and FAM134B, we discovered that ATZ coimmunoprecipitates with the p24-family members TMP21 and TMED9. This contrasts with wild type alpha1-antitrypsin, which did not coimmunoprecipitate with FAM134B, calnexin or the p24-family members. Live-cell imaging revealed that ATZ and the p24-family members traffic together from the ER to lysosomes. Using chemical inhibitors to block ER exit or autophagy, we demonstrated that p24-family members and ATZ co-accumulate at SEC24C marked ER-exit sites or in ER-derived compartments, respectively. Furthermore, depletion of SEC24C, TMP21, or TMED9 inhibited lysosomal trafficking of ATZ and resulted in the increase of intracellular ATZ levels. Conversely, overexpression of these p24-family members resulted in the reduction of ATZ levels. Intriguingly, the p24-family members coimmunoprecipitate with ATZ, FAM134B, and SEC24C. Thus, we propose a model in which the p24-family functions in an adaptor complex linking SEC24C with the ERLAD machinery for the clearance of ATZ.
Collapse
Affiliation(s)
| | - Debashree Mitra
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Sudhanshu Abishek
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Richa Beher
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | | |
Collapse
|
11
|
Li T, Yang F, Heng Y, Zhou S, Wang G, Wang J, Wang J, Chen X, Yao ZP, Wu Z, Guo Y. TMED10 mediates the trafficking of insulin-like growth factor 2 along the secretory pathway for myoblast differentiation. Proc Natl Acad Sci U S A 2023; 120:e2215285120. [PMID: 37931110 PMCID: PMC10655563 DOI: 10.1073/pnas.2215285120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/02/2023] [Indexed: 11/08/2023] Open
Abstract
The insulin-like growth factor 2 (IGF2) plays critical roles in cell proliferation, migration, differentiation, and survival. Despite its importance, the molecular mechanisms mediating the trafficking of IGF2 along the secretory pathway remain unclear. Here, we utilized a Retention Using Selective Hook system to analyze molecular mechanisms that regulate the secretion of IGF2. We found that a type I transmembrane protein, TMED10, is essential for the secretion of IGF2 and for differentiation of mouse myoblast C2C12 cells. Further analyses indicate that the residues 112-140 in IGF2 are important for the secretion of IGF2 and these residues directly interact with the GOLD domain of TMED10. We then reconstituted the release of IGF2 into COPII vesicles. This assay suggests that TMED10 mediates the packaging of IGF2 into COPII vesicles to be efficiently delivered to the Golgi. Moreover, TMED10 also mediates ER export of TGN-localized cargo receptor, sortilin, which subsequently mediates TGN export of IGF2. These analyses indicate that TMED10 is critical for IGF2 secretion by directly regulating ER export and indirectly regulating TGN export of IGF2, providing insights into trafficking of IGF2 for myoblast differentiation.
Collapse
Affiliation(s)
- Tiantian Li
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Feng Yang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Youshan Heng
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shaopu Zhou
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Gang Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jianying Wang
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Research Centre for Chinese Medicine Innovation, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jinhui Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xianwei Chen
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhong-Ping Yao
- State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food, Research Centre for Chinese Medicine Innovation, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) and Shenzhen Key Laboratory of Food Biological Safety Control, Hong Kong Polytechnic University, Shenzhen Research Institute, Shenzhen 518057, China
| | - Zhenguo Wu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yusong Guo
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong University of Science and Technology, Shenzhen Research Institute, Shenzhen 518057, China
- Thrust of Bioscience and Biomedical Engineering, Hong Kong University of Science and Technology, Guangzhou 511453, China
| |
Collapse
|
12
|
Holm JEJ, Soares SG, Symmons MF, Huddin AS, Moncrieffe MC, Gay NJ. Anterograde trafficking of Toll-like receptors requires the cargo sorting adaptors TMED-2 and 7. Traffic 2023; 24:508-521. [PMID: 37491993 PMCID: PMC10946956 DOI: 10.1111/tra.12912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/15/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
Toll-Like Receptors (TLRs) play a pivotal role in immunity by recognising conserved structural features of pathogens and initiating the innate immune response. TLR signalling is subject to complex regulation that remains poorly understood. Here we show that two small type I transmembrane receptors, TMED2 and 7, that function as cargo sorting adaptors in the early secretory pathway are required for transport of TLRs from the ER to Golgi. Protein interaction studies reveal that TMED7 interacts with TLR2, TLR4 and TLR5 but not with TLR3 and TLR9. On the other hand, TMED2 interacts with TLR2, TLR4 and TLR3. Dominant negative forms of TMED7 suppress the export of cell surface TLRs from the ER to the Golgi. By contrast TMED2 is required for the ER-export of both plasma membrane and endosomal TLRs. Together, these findings suggest that association of TMED2 and TMED7 with TLRs facilitates anterograde transport from the ER to the Golgi.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicholas J. Gay
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
13
|
Tao Z, Yang D, Ni R. Tmed10 deficiency results in impaired exocrine pancreatic differentiation in zebrafish larvae. Dev Biol 2023; 503:43-52. [PMID: 37597605 DOI: 10.1016/j.ydbio.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Transmembrane p24 trafficking protein 10 (TMED10) is a conserved vesicle trafficking protein. It is dysregulated in Alzheimer disease and plays a pivotal role in the pathogenesis of Alzheimer disease. In addition to the brain, TMED10 is highly expressed in the exocrine pancreas; however, its biological functions and underlying mechanisms remain largely unknown. We studied reduced Tmed10 in zebrafish embryos by morpholino oligonucleotide knockdown and CRISPR-Cas9 mutagenesis. Tmed10-deficient embryos showed extensive loss of acinar mass and impaired acinar differentiation. TMED10 has been reported to have an inhibitory effect on γ-secretase. As one of the substrates of γ-secretase, membrane-bound β-catenin was significantly reduced in Tmed10-deficient embryos. Increased γ-secretase activity in wild-type embryos resulted in a phenotype similar to that of tmed10 mutants. And the mutant phenotype could be rescued by treatment with the γ-secretase inhibitor, N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-s-phenylglycinet-butyl ester (DAPT). In addition, the reduced membrane-bound β-catenin was accompanied with up-regulated β-catenin target genes in Tmed10-deficient embryos. Overexpression of β-catenin signaling inhibitor Dickkopf-1 (DKK-1) could rescue the exocrine pancreas defects. Taken together, our study reveals that Tmed10 regulates exocrine pancreatic differentiation through γ-secretase. Reduced membrane-bound β-catenin, accompanied with hyperactivation of β-catenin signaling, is an important cause of exocrine pancreas defects in Tmed10-deficient embryos. Our study reaffirms the importance of appropriate β-catenin signaling in exocrine pancreas development. These findings may provide a theoretical basis for the development of treatment strategies for TMED10-related diseases.
Collapse
Affiliation(s)
- Zewen Tao
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China
| | - Di Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715, Chongqing, China.
| |
Collapse
|
14
|
Xu X, Lambert NA, Wu G. Sequence-directed concentration of G protein-coupled receptors in COPII vesicles. iScience 2023; 26:107969. [PMID: 37810244 PMCID: PMC10551652 DOI: 10.1016/j.isci.2023.107969] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/02/2023] [Accepted: 09/15/2023] [Indexed: 10/10/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest superfamily of plasma membrane signaling proteins. However, virtually nothing is known about their recruitment to COPII vesicles for forward delivery after synthesis in the endoplasmic reticulum (ER). Here, we demonstrate that some GPCRs are highly concentrated at ER exit sites (ERES) before COPII budding. Angiotensin II type 2 receptor (AT2R) and CXCR4 concentration are directed by a di-acidic motif and a 9-residue domain, respectively, and these motifs also control receptor ER-Golgi traffic. We further show that AT2R interacts with Sar1 GTPase and that distinct GPCRs have different ER-Golgi transport rates via COPII which is independent of their concentration at ERES. Collectively, these data demonstrate that GPCRs can be actively captured by COPII via specific motifs and direct interaction with COPII components that in turn affects their export dynamics, and provide important insights into COPII targeting and forward trafficking of nascent GPCRs.
Collapse
Affiliation(s)
- Xin Xu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nevin A. Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Abi Nahed R, Safwan-Zaiter H, Gemy K, Lyko C, Boudaud M, Desseux M, Marquette C, Barjat T, Alfaidy N, Benharouga M. The Multifaceted Functions of Prion Protein (PrP C) in Cancer. Cancers (Basel) 2023; 15:4982. [PMID: 37894349 PMCID: PMC10605613 DOI: 10.3390/cancers15204982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The cellular prion protein (PrPC) is a glycoprotein anchored to the cell surface by glycosylphosphatidylinositol (GPI). PrPC is expressed both in the brain and in peripheral tissues. Investigations on PrPC's functions revealed its direct involvement in neurodegenerative and prion diseases, as well as in various physiological processes such as anti-oxidative functions, copper homeostasis, trans-membrane signaling, and cell adhesion. Recent findings have revealed the ectopic expression of PrPC in various cancers including gastric, melanoma, breast, colorectal, pancreatic, as well as rare cancers, where PrPC promotes cellular migration and invasion, tumor growth, and metastasis. Through its downstream signaling, PrPC has also been reported to be involved in resistance to chemotherapy and tumor cell apoptosis. This review summarizes the variance of expression of PrPC in different types of cancers and discusses its roles in their development and progression, as well as its use as a potential target to treat such cancers.
Collapse
Affiliation(s)
- Roland Abi Nahed
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Hasan Safwan-Zaiter
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Kevin Gemy
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Camille Lyko
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Mélanie Boudaud
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Morgane Desseux
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Christel Marquette
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Tiphaine Barjat
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Nadia Alfaidy
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Mohamed Benharouga
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| |
Collapse
|
16
|
Navarro KG, Chamberlin HM. Genetic characterization of C. elegans TMED genes. Dev Dyn 2023; 252:1149-1161. [PMID: 37204056 PMCID: PMC10524739 DOI: 10.1002/dvdy.601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND p24/transmembrane Emp24 domain (TMED) proteins are a set of evolutionarily conserved, single pass transmembrane proteins that have been shown to facilitate protein secretion and selection of cargo proteins to transport vesicles in the cellular secretion pathway. However, their functions in animal development are incompletely understood. RESULTS The C. elegans genome encodes eight identified TMED genes, with at least one member from each defined subfamily (α, β, γ, δ). TMED gene mutants exhibit a shared set of defects in embryonic viability, animal movement, and vulval morphology. Two γ subfamily genes, tmed-1 and tmed-3, exhibit the ability to compensate for each other, as defects in movement and vulva morphology are only apparent in double mutants. TMED mutants also exhibit a delay in breakdown of basement membrane during vulva development. CONCLUSIONS The results establish a genetic and experimental framework for the study of TMED gene function in C. elegans, and argue that a functional protein from each subfamily is important for a shared set of developmental processes. A specific function for TMED genes is to facilitate breakdown of the basement membrane between the somatic gonad and vulval epithelial cells, suggesting a role for TMED proteins in tissue reorganization during animal development.
Collapse
|
17
|
Almeida SM, Ivantsiv S, Niibori R, Dunham WH, Green BA, Zhao L, Gingras AC, Cordes SP. An interaction between OTULIN and SCRIB uncovers roles for linear ubiquitination in planar cell polarity. Dis Model Mech 2023; 16:dmm049762. [PMID: 37589075 PMCID: PMC10445738 DOI: 10.1242/dmm.049762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 07/17/2023] [Indexed: 08/18/2023] Open
Abstract
Planar cell polarity (PCP) plays critical roles in developmental and homeostatic processes. Membrane presentation of PCP complexes containing Van Gogh-like (VANGL) transmembrane proteins is central to PCP and can be directed by the scaffold protein scribble (SCRIB). The role atypical linear ubiquitin (Met1-Ub) chains might play in PCP is unknown. Here, HEK293 cell-based interactomic analyses of the Met1-Ub deubiquitinase OTULIN revealed that OTULIN can interact with SCRIB. Moreover, Met1-Ub chains associated with VANGL2 and PRICKLE1, but not SCRIB, can direct VANGL2 surface presentation. Mouse embryos lacking Otulin showed variable neural tube malformations, including rare open neural tubes, a deficit associated with PCP disruption in mice. In Madin-Darby canine kidney cells, in which the enrichment of VANGL2-GFP proteins at cell-cell contacts represents activated PCP complexes, endogenous OTULIN was recruited to these sites. In the human MDA-MB-231 breast cancer cell model, OTULIN loss caused deficits in Wnt5a-induced filopodia extension and trafficking of transfected HA-VANGL2. Taken together, these findings support a role for linear (de)ubiquitination in PCP signaling. The association of Met1-Ub chains with PCP complex components offers new opportunities for integrating PCP signaling with OTULIN-dependent immune and inflammatory pathways.
Collapse
Affiliation(s)
- Stephanie M. Almeida
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sofiia Ivantsiv
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rieko Niibori
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
| | - Wade H. Dunham
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Brooke A. Green
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liang Zhao
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabine P. Cordes
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
18
|
Aguilera-Romero A, Lucena R, Sabido-Bozo S, Muñiz M. Impact of sphingolipids on protein membrane trafficking. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159334. [PMID: 37201864 DOI: 10.1016/j.bbalip.2023.159334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/28/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Membrane trafficking is essential to maintain the spatiotemporal control of protein and lipid distribution within membrane systems of eukaryotic cells. To achieve their functional destination proteins are sorted and transported into lipid carriers that construct the secretory and endocytic pathways. It is an emerging theme that lipid diversity might exist in part to ensure the homeostasis of these pathways. Sphingolipids, a chemical diverse type of lipids with special physicochemical characteristics have been implicated in the selective transport of proteins. In this review, we will discuss current knowledge about how sphingolipids modulate protein trafficking through the endomembrane systems to guarantee that proteins reach their functional destination and the proposed underlying mechanisms.
Collapse
Affiliation(s)
- Auxiliadora Aguilera-Romero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| | - Rafael Lucena
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Susana Sabido-Bozo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| |
Collapse
|
19
|
Tang VT, Ginsburg D. Cargo selection in endoplasmic reticulum-to-Golgi transport and relevant diseases. J Clin Invest 2023; 133:163838. [PMID: 36594468 PMCID: PMC9797344 DOI: 10.1172/jci163838] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Most proteins destined for the extracellular space or various intracellular compartments must traverse the intracellular secretory pathway. The first step is the recruitment and transport of cargoes from the endoplasmic reticulum (ER) lumen to the Golgi apparatus by coat protein complex II (COPII), consisting of five core proteins. Additional ER transmembrane proteins that aid cargo recruitment are referred to as cargo receptors. Gene duplication events have resulted in multiple COPII paralogs present in the mammalian genome. Here, we review the functions of each COPII protein, human disorders associated with each paralog, and evidence for functional conservation between paralogs. We also provide a summary of current knowledge regarding two prototypical cargo receptors in mammals, LMAN1 and SURF4, and their roles in human health and disease.
Collapse
Affiliation(s)
- Vi T. Tang
- Department of Molecular and Integrative Physiology,,Life Sciences Institute
| | - David Ginsburg
- Life Sciences Institute,,Department of Internal Medicine,,Department of Human Genetics,,Department of Pediatrics and Communicable Diseases, and,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Ostwaldt F, Los B, Heyd F. In silico analysis of alternative splicing events implicated in intracellular trafficking during B-lymphocyte differentiation. Front Immunol 2022; 13:1030409. [DOI: 10.3389/fimmu.2022.1030409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
There are multiple regulatory layers that control intracellular trafficking and protein secretion, ranging from transcriptional to posttranslational mechanisms. Finely regulated trafficking and secretion is especially important for lymphocytes during activation and differentiation, as the quantity of secretory cargo increases once the activated cells start to produce and secrete large amounts of cytokines, cytotoxins, or antibodies. However, how the secretory machinery dynamically adapts its efficiency and specificity in general and specifically in lymphocytes remains incompletely understood. Here we present a systematic bioinformatics analysis to address RNA-based mechanisms that control intracellular trafficking and protein secretion during B-lymphocyte activation, and differentiation, with a focus on alternative splicing. Our in silico analyses suggest that alternative splicing has a substantial impact on the dynamic adaptation of intracellular traffic and protein secretion in different B cell subtypes, pointing to another regulatory layer to the control of lymphocyte function during activation and differentiation. Furthermore, we suggest that NERF/ELF2 controls the expression of some COPII-related genes in a cell type-specific manner. In addition, T cells and B cells appear to use different adaptive strategies to adjust their secretory machineries during the generation of effector and memory cells, with antibody secreting B cell specifically increasing the expression of components of the early secretory pathway. Together, our data provide hypotheses how cell type-specific regulation of the trafficking machinery during immune cell activation and differentiation is controlled that can now be tested in wet lab experiments.
Collapse
|
21
|
New Players in Neuronal Iron Homeostasis: Insights from CRISPRi Studies. Antioxidants (Basel) 2022; 11:antiox11091807. [PMID: 36139881 PMCID: PMC9495848 DOI: 10.3390/antiox11091807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Selective regional iron accumulation is a hallmark of several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. The underlying mechanisms of neuronal iron dyshomeostasis have been studied, mainly in a gene-by-gene approach. However, recent high-content phenotypic screens using CRISPR/Cas9-based gene perturbations allow for the identification of new pathways that contribute to iron accumulation in neuronal cells. Herein, we perform a bioinformatic analysis of a CRISPR-based screening of lysosomal iron accumulation and the functional genomics of human neurons derived from induced pluripotent stem cells (iPSCs). Consistent with previous studies, we identified mitochondrial electron transport chain dysfunction as one of the main mechanisms triggering iron accumulation, although we substantially expanded the gene set causing this phenomenon, encompassing mitochondrial complexes I to IV, several associated assembly factors, and coenzyme Q biosynthetic enzymes. Similarly, the loss of numerous genes participating through the complete macroautophagic process elicit iron accumulation. As a novelty, we found that the impaired synthesis of glycophosphatidylinositol (GPI) and GPI-anchored protein trafficking also trigger iron accumulation in a cell-autonomous manner. Finally, the loss of critical components of the iron transporters trafficking machinery, including MON2 and PD-associated gene VPS35, also contribute to increased neuronal levels. Our analysis suggests that neuronal iron accumulation can arise from the dysfunction of an expanded, previously uncharacterized array of molecular pathways.
Collapse
|
22
|
Gomez-Navarro N, Maldutyte J, Poljak K, Peak-Chew SY, Orme J, Bisnett BJ, Lamb CH, Boyce M, Gianni D, Miller EA. Selective inhibition of protein secretion by abrogating receptor-coat interactions during ER export. Proc Natl Acad Sci U S A 2022; 119:e2202080119. [PMID: 35901214 PMCID: PMC9351455 DOI: 10.1073/pnas.2202080119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 01/03/2023] Open
Abstract
Protein secretion is an essential process that drives cell growth, movement, and communication. Protein traffic within the secretory pathway occurs via transport intermediates that bud from one compartment and fuse with a downstream compartment to deliver their contents. Here, we explore the possibility that protein secretion can be selectively inhibited by perturbing protein-protein interactions that drive capture into transport vesicles. Human proprotein convertase subtilisin/kexin type 9 (PCSK9) is a determinant of cholesterol metabolism whose secretion is mediated by a specific cargo adaptor protein, SEC24A. We map a series of protein-protein interactions between PCSK9, its endoplasmic reticulum (ER) export receptor SURF4, and SEC24A that mediate secretion of PCSK9. We show that the interaction between SURF4 and SEC24A can be inhibited by 4-phenylbutyrate (4-PBA), a small molecule that occludes a cargo-binding domain of SEC24. This inhibition reduces secretion of PCSK9 and additional SURF4 clients that we identify by mass spectrometry, leaving other secreted cargoes unaffected. We propose that selective small-molecule inhibition of cargo recognition by SEC24 is a potential therapeutic intervention for atherosclerosis and other diseases that are modulated by secreted proteins.
Collapse
Affiliation(s)
- Natalia Gomez-Navarro
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Julija Maldutyte
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Kristina Poljak
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Sew-Yeu Peak-Chew
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| | - Jonathon Orme
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Brittany J. Bisnett
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Caitlin H. Lamb
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine, Durham, NC27710, USA
| | - Davide Gianni
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, United Kingdom
| | - Elizabeth A. Miller
- Cell Biology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom
| |
Collapse
|
23
|
Dimou S, Dionysopoulou M, Sagia GM, Diallinas G. Golgi-Bypass Is a Major Unconventional Route for Translocation to the Plasma Membrane of Non-Apical Membrane Cargoes in Aspergillus nidulans. Front Cell Dev Biol 2022; 10:852028. [PMID: 35465316 PMCID: PMC9021693 DOI: 10.3389/fcell.2022.852028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Nutrient transporters have been shown to translocate to the plasma membrane (PM) of the filamentous fungus Aspergillus nidulans via an unconventional trafficking route that bypasses the Golgi. This finding strongly suggests the existence of distinct COPII vesicle subpopulations, one following Golgi-dependent conventional secretion and the other directed towards the PM. Here, we address whether Golgi-bypass concerns cargoes other than nutrient transporters and whether Golgi-bypass is related to cargo structure, size, abundance, physiological function, or polar vs. non-polar distribution in the PM. To address these questions, we followed the dynamic subcellular localization of two selected membrane cargoes differing in several of the aforementioned aspects. These are the proton-pump ATPase PmaA and the PalI pH signaling component. Our results show that neosynthesized PmaA and PalI are translocated to the PM via Golgi-bypass, similar to nutrient transporters. In addition, we showed that the COPII-dependent exit of PmaA from the ER requires the alternative COPII coat subunit LstA, rather than Sec24, whereas PalI requires the ER cargo adaptor Erv14. These findings strengthen the evidence of distinct cargo-specific COPII subpopulations and extend the concept of Golgi-independent biogenesis to essential transmembrane proteins, other than nutrient transporters. Overall, our findings point to the idea that Golgi-bypass might not constitute a fungal-specific peculiarity, but rather a novel major and cargo-specific sorting route in eukaryotic cells that has been largely ignored.
Collapse
Affiliation(s)
- Sofia Dimou
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, Athens, Greece
| | - Mariangela Dionysopoulou
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, Athens, Greece
| | - Georgia Maria Sagia
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, Athens, Greece
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, Athens, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- *Correspondence: George Diallinas,
| |
Collapse
|
24
|
Mendes LFS, Costa-Filho AJ. A gold revision of the Golgi Dynamics (GOLD) domain structure and associated cell functionalities. FEBS Lett 2022; 596:973-990. [PMID: 35099811 DOI: 10.1002/1873-3468.14300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/06/2022]
Abstract
The classical secretory pathway is the key membrane-based delivery system in eukaryotic cells. Several families of proteins involved in the secretory pathway, with functionalities going from cargo sorting receptors to the maintenance and dynamics of secretory organelles, share soluble globular domains predicted to mediate protein-protein interactions. One of them is "Golgi Dynamics" (GOLD) domain, named after its strong association with the Golgi apparatus. There are many GOLD-containing protein families, such as the Transmembrane emp24 domain-containing proteins (TMED/p24 family), animal SEC14-like proteins, Human Golgi resident protein ACBD3, a splice variant of TICAM2 called TRAM with GOLD domain and FYCO1. Here, we critically review the state-of-the-art knowledge of the structures and functions of the main representatives of GOLD-containing proteins in vertebrates. We provide the first unified description of the GOLD domain structure across different families since the first high-resolution structure was determined. With a brand-new update on the definition of the GOLD domain, we also discuss how its tertiary structure fits the β-sandwich-like fold map and give exciting new directions for forthcoming studies.
Collapse
Affiliation(s)
- Luis Felipe S Mendes
- Laboratório de Biofísica Molecular, Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Antonio J Costa-Filho
- Laboratório de Biofísica Molecular, Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
25
|
Sargeant J, Seiler DK, Costain T, Madreiter-Sokolowski CT, Gordon DE, Peden AA, Malli R, Graier WF, Hay JC. ALG-2 and peflin regulate COPII targeting and secretion in response to calcium signaling. J Biol Chem 2021; 297:101393. [PMID: 34762908 PMCID: PMC8671942 DOI: 10.1016/j.jbc.2021.101393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 02/05/2023] Open
Abstract
ER-to-Golgi transport is the first step in the constitutive secretory pathway, which, unlike regulated secretion, is believed to proceed nonstop independent of Ca2+ flux. However, here we demonstrate that penta-EF hand (PEF) proteins ALG-2 and peflin constitute a hetero-bifunctional COPII regulator that responds to Ca2+ signaling by adopting one of several distinct activity states. Functionally, these states can adjust the rate of ER export of COPII-sorted cargos up or down by ∼50%. We found that at steady-state Ca2+, ALG-2/peflin hetero-complexes bind to ER exit sites (ERES) through the ALG-2 subunit to confer a low, buffered secretion rate, while peflin-lacking ALG-2 complexes markedly stimulate secretion. Upon Ca2+ signaling, ALG-2 complexes lacking peflin can either increase or decrease the secretion rate depending on signaling intensity and duration-phenomena that could contribute to cellular growth and intercellular communication following secretory increases or protection from excitotoxicity and infection following decreases. In epithelial normal rat kidney (NRK) cells, the Ca2+-mobilizing agonist ATP causes ALG-2 to depress ER export, while in neuroendocrine PC12 cells, Ca2+ mobilization by ATP results in ALG-2-dependent enhancement of secretion. Furthermore, distinct Ca2+ signaling patterns in NRK cells produce opposing ALG-2-dependent effects on secretion. Mechanistically, ALG-2-dependent depression of secretion involves decreased levels of the COPII outer shell and increased peflin targeting to ERES, while ALG-2-dependent enhancement of secretion involves increased COPII outer shell and decreased peflin at ERES. These data provide insights into how PEF protein dynamics affect secretion of important physiological cargoes such as collagen I and significantly impact ER stress.
Collapse
Affiliation(s)
- John Sargeant
- Division of Biological Sciences, Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana, USA
| | - Danette Kowal Seiler
- Division of Biological Sciences, Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana, USA
| | - Tucker Costain
- Division of Biological Sciences, Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana, USA
| | | | - David E Gordon
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Andrew A Peden
- Department of Biomedical Science and Centre for Membrane Interactions and Dynamics, The University of Sheffield, Sheffield, United Kingdom
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Jesse C Hay
- Division of Biological Sciences, Center for Structural and Functional Neuroscience, University of Montana, Missoula, Montana, USA.
| |
Collapse
|
26
|
Adams EJ, Khoriaty R, Kiseleva A, Cleuren ACA, Tomberg K, van der Ent MA, Gergics P, Tang VT, Zhu G, Hoenerhoff MJ, O'Shea KS, Saunders TL, Ginsburg D. Murine SEC24D can substitute functionally for SEC24C during embryonic development. Sci Rep 2021; 11:21100. [PMID: 34702932 PMCID: PMC8548507 DOI: 10.1038/s41598-021-00579-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022] Open
Abstract
The COPII component SEC24 mediates the recruitment of transmembrane cargos or cargo adaptors into newly forming COPII vesicles on the ER membrane. Mammalian genomes encode four Sec24 paralogs (Sec24a-d), with two subfamilies based on sequence homology (SEC24A/B and C/D), though little is known about their comparative functions and cargo-specificities. Complete deficiency for Sec24d results in very early embryonic lethality in mice (before the 8 cell stage), with later embryonic lethality (E7.5) observed in Sec24c null mice. To test the potential overlap in function between SEC24C/D, we employed dual recombinase mediated cassette exchange to generate a Sec24cc-d allele, in which the C-terminal 90% of SEC24C has been replaced by SEC24D coding sequence. In contrast to the embryonic lethality at E7.5 of SEC24C-deficiency, Sec24cc-d/c-d pups survive to term, though dying shortly after birth. Sec24cc-d/c-d pups are smaller in size, but exhibit no other obvious developmental abnormality by pathologic evaluation. These results suggest that tissue-specific and/or stage-specific expression of the Sec24c/d genes rather than differences in cargo export function explain the early embryonic requirements for SEC24C and SEC24D.
Collapse
Affiliation(s)
- Elizabeth J Adams
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Constellation Pharmaceuticals, Cambridge, MA, 02142, USA
| | - Rami Khoriaty
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Univeristy of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA.
| | - Anna Kiseleva
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Audrey C A Cleuren
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kärt Tomberg
- Departement of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Peter Gergics
- Departement of Human Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Vi T Tang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Guojing Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mark J Hoenerhoff
- In Vivo Animal Core, Unit of Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David Ginsburg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, 48109, USA.
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
27
|
ER Morphology in the Pathogenesis of Hereditary Spastic Paraplegia. Cells 2021; 10:cells10112870. [PMID: 34831093 PMCID: PMC8616106 DOI: 10.3390/cells10112870] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) is the most abundant and widespread organelle in cells. Its peculiar membrane architecture, formed by an intricate network of tubules and cisternae, is critical to its multifaceted function. Regulation of ER morphology is coordinated by a few ER-specific membrane proteins and is thought to be particularly important in neurons, where organized ER membranes are found even in the most distant neurite terminals. Mutation of ER-shaping proteins has been implicated in the neurodegenerative disease hereditary spastic paraplegia (HSP). In this review we discuss the involvement of these proteins in the pathogenesis of HSP, focusing on the experimental evidence linking their molecular function to disease onset. Although the precise biochemical activity of some ER-related HSP proteins has been elucidated, the pathological mechanism underlying ER-linked HSP is still undetermined and needs to be further investigated.
Collapse
|
28
|
Tashima Y, Hirata T, Maeda Y, Murakami Y, Kinoshita T. Differential use of p24 family members as cargo receptors for the transport of glycosylphosphatidylinositol-anchored proteins and Wnt1. J Biochem 2021; 171:75-83. [PMID: 34647572 DOI: 10.1093/jb/mvab108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/12/2021] [Indexed: 02/01/2023] Open
Abstract
Complexes of p24 proteins act as cargo receptors for the transport of COPII vesicles from the endoplasmic reticulum. The major cargos of p24 complexes are hydrophilic proteins tethered to the endoplasmic reticulum membrane via a covalently attached glycosylphosphatidylinositol (GPI) or fatty acid. Each p24 complex is known to contain members from all four p24 subfamilies (p24α, p24β, p24γ, and p24δ). However, it remains unclear how the cargo specificities of p24 complexes are influenced by member stoichiometry. Here, we report the subunit compositions of mammalian p24 complexes involved in the transport of GPI-anchored proteins and Wnt1. We show that at least one p24α is required for the formation of p24 complexes, and that a p24 complex consisting of p24α2, p24β1, p24γ2, and p24δ1 is required for the efficient transport of GPI-anchored proteins. On the other hand, a p24 complex containing p24α2, p24α3, p24β1, p24γ, and p24δ1 is involved in the transport of Wnt1. Further, interactions between p24α2 and p24α3 are critical for Wnt1 transport. Thus, p24α and p24γ subfamily members are important for cargo selectivity. Lastly, our data fit with an octamer, rather than a tetramer, model of p24 complexes, where each complex consists of two proteins from each p24 subfamily.
Collapse
Affiliation(s)
- Yuko Tashima
- Research Institute for Microbial Diseases, and + WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan.,Current Address: Department of Molecular & Cellular Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Tetsuya Hirata
- Research Institute for Microbial Diseases, and + WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan.,Current Address: Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu 501-1193, Japan
| | - Yusuke Maeda
- Research Institute for Microbial Diseases, and + WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiko Murakami
- Research Institute for Microbial Diseases, and + WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, and + WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
29
|
Chatterjee S, Choi AJ, Frankel G. A systematic review of Sec24 cargo interactome. Traffic 2021; 22:412-424. [PMID: 34533884 DOI: 10.1111/tra.12817] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/27/2021] [Accepted: 09/13/2021] [Indexed: 01/10/2023]
Abstract
Endoplasmic reticulum (ER)-to-Golgi trafficking is an essential and highly conserved cellular process. The coat protein complex-II (COPII) arm of the trafficking machinery incorporates a wide array of cargo proteins into vesicles through direct or indirect interactions with Sec24, the principal subunit of the COPII coat. Approximately one-third of all mammalian proteins rely on the COPII-mediated secretory pathway for membrane insertion or secretion. There are four mammalian Sec24 paralogs and three yeast Sec24 paralogs with emerging evidence of paralog-specific cargo interaction motifs. Furthermore, individual paralogs also differ in their affinity for a subset of sorting motifs present on cargo proteins. As with many aspects of protein trafficking, we lack a systematic and thorough understanding of the interaction of Sec24 with cargoes. This systematic review focuses on the current knowledge of cargo binding to both yeast and mammalian Sec24 paralogs and their ER export motifs. The analyses show that Sec24 paralog specificity of cargo (and cargo receptors) range from exclusive paralog dependence or preference to partial redundancy. We also discuss how the Sec24 secretion system is hijacked by viral (eg, VSV-G, Hepatitis B envelope protein) and bacterial (eg, the enteropathogenic Escherichia coli type III secretion system effector NleA/EspI) pathogens.
Collapse
Affiliation(s)
- Sharanya Chatterjee
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Ana Jeemin Choi
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK
| |
Collapse
|
30
|
Shijie L, Zhen P, Kang Q, Hua G, Qingcheng Y, Dongdong C. Deregulation of CLTC interacts with TFG, facilitating osteosarcoma via the TGF-beta and AKT/mTOR signaling pathways. Clin Transl Med 2021; 11:e377. [PMID: 34185412 PMCID: PMC8214859 DOI: 10.1002/ctm2.377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 01/05/2023] Open
Abstract
Although the treatment of osteosarcoma has improved, the overall survival rate of this common type of osseous malignancies has not changed for four decades. Thus, new targets for better therapeutic regimens are urgently needed. In this study, we found that high expression of clathrin heavy chain (CLTC) was an independent prognostic factor for tumor-free survival (HzR, 3.049; 95% CI, 1.476-6.301) and overall survival (HzR, 2.469; 95% CI, 1.005-6.067) of patients with osteosarcoma. Down-regulation of CLTC resulted in tumor-suppressive effects in vitro and in vivo. Moreover, we found that CLTC was transcriptionally regulated by a transcription factor-specificity protein 1 (SP1), which binds to the CLTC promoter at the -320 to -314-nt and +167 to +173-nt loci. Mechanistic investigations further revealed that CLTC elicited its pro-tumor effects by directly binding to and stabilizing trafficking from the endoplasmic reticulum to the Golgi regulator (TFG). Importantly, overexpression of TFG rescued both the tumor-suppressive effect and inhibition of the TGF-β and AKT/mTOR pathways caused by CLTC down-regulation, which indicated that the activity of CLTC was TFG-dependent. Immunohistochemistry analysis confirmed that CLTC expression was positively correlated with TFG expression. These findings collectively highlight CLTC as a new prognostic biomarker for patients with osteosarcoma, and the interruption of the SP1/CLTC/TFG axis may serve as a novel therapeutic strategy for osteosarcoma.
Collapse
Affiliation(s)
- Li Shijie
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Pan Zhen
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Qin Kang
- Department of Trauma and Reconstructive SurgeryRWTH Aachen University HospitalAachenGermany
| | - Guo Hua
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yang Qingcheng
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Cheng Dongdong
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| |
Collapse
|
31
|
Tabasi M, Nombela I, Janssens J, Lahousse AP, Christ F, Debyser Z. Role of Transportin-SR2 in HIV-1 Nuclear Import. Viruses 2021; 13:829. [PMID: 34064404 PMCID: PMC8147801 DOI: 10.3390/v13050829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022] Open
Abstract
The HIV replication cycle depends on the interaction of viral proteins with proteins of the host. Unraveling host-pathogen interactions during the infection is of great importance for understanding the pathogenesis and the development of antiviral therapies. To date HIV uncoating and nuclear import are the most debated steps of the HIV-1 replication cycle. Despite numerous studies during past decades, there is still much controversy with respect to the identity and the role of viral and host factors involved in these processes. In this review, we provide a comprehensive overview on the role of transportin-SR2 as a host cell factor during active nuclear transport.
Collapse
Affiliation(s)
| | | | | | | | | | - Zeger Debyser
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Flanders, Belgium; (M.T.); (I.N.); (J.J.); (A.P.L.); (F.C.)
| |
Collapse
|
32
|
Bozorgmehr N, Okoye I, Oyegbami O, Xu L, Fontaine A, Cox-Kennett N, Larratt LM, Hnatiuk M, Fagarasanu A, Brandwein J, Peters AC, Elahi S. Expanded antigen-experienced CD160 +CD8 +effector T cells exhibit impaired effector functions in chronic lymphocytic leukemia. J Immunother Cancer 2021; 9:jitc-2020-002189. [PMID: 33931471 PMCID: PMC8098955 DOI: 10.1136/jitc-2020-002189] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background T cell exhaustion compromises antitumor immunity, and a sustained elevation of co-inhibitory receptors is a hallmark of T cell exhaustion in solid tumors. Similarly, upregulation of co-inhibitory receptors has been reported in T cells in hematological cancers such as chronic lymphocytic leukemia (CLL). However, the role of CD160, a glycosylphosphatidylinositol-anchored protein, as one of these co-inhibitory receptors has been contradictory in T cell function. Therefore, we decided to elucidate how CD160 expression and/or co-expression with other co-inhibitory receptors influence T cell effector functions in patients with CLL. Methods We studied 56 patients with CLL and 25 age-matched and sex-matched healthy controls in this study. The expression of different co-inhibitory receptors was analyzed in T cells obtained from the peripheral blood or the bone marrow. Also, we quantified the properties of extracellular vesicles (EVs) in the plasma of patients with CLL versus healthy controls. Finally, we measured 29 different cytokines, chemokines or other biomarkers in the plasma specimens of patients with CLL and healthy controls. Results We found that CD160 was the most upregulated co-inhibitory receptor in patients with CLL. Its expression was associated with an exhausted T cell phenotype. CD160+CD8+ T cells were highly antigen-experienced/effector T cells, while CD160+CD4+ T cells were more heterogeneous. In particular, we identified EVs as a source of CD160 in the plasma of patients with CLL that can be taken up by T cells. Moreover, we observed a dominantly proinflammatory cytokine profile in the plasma of patients with CLL. In particular, interleukin-16 (IL-16) was highly elevated and correlated with the advanced clinical stage (Rai). Furthermore, we observed that the incubation of T cells with IL-16 results in the upregulation of CD160. Conclusions Our study provides a novel insight into the influence of CD160 expression/co-expression with other co-inhibitory receptors in T cell effector functions in patients with CLL. Besides, IL-16-mediated upregulation of CD160 expression in T cells highlights the importance of IL-16/CD160 as potential immunotherapy targets in patients with CLL. Therefore, our findings propose a significant role for CD160 in T cell exhaustion in patients with CLL.
Collapse
Affiliation(s)
- Najmeh Bozorgmehr
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Isobel Okoye
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Olaide Oyegbami
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lai Xu
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amelie Fontaine
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nanette Cox-Kennett
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Loree M Larratt
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Hnatiuk
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andrei Fagarasanu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph Brandwein
- Division of Hematology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anthea C Peters
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shokrollah Elahi
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada .,Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
33
|
Malekshahi Z, Bernklau S, Schiela B, Koske I, Banki Z, Stiasny K, Harris CL, Würzner R, Stoiber H. Incorporation of CD55 into the Zika Viral Envelope Contributes to Its Stability against Human Complement. Viruses 2021; 13:v13030510. [PMID: 33808725 PMCID: PMC8003375 DOI: 10.3390/v13030510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/25/2022] Open
Abstract
The rapid spread of the virus in Latin America and the association of the infection with microcephaly in newborns or Guillain–Barré Syndrome in adults prompted the WHO to declare the Zika virus (ZIKV) epidemic to be an international public health emergency in 2016. As the virus was first discovered in monkeys and is spread not only by mosquitos but also from human to human, we investigated the stability to the human complement of ZIKV derived from mosquito (ZIKVInsect), monkey (ZIKVVero), or human cells (ZIKVA549 and ZIKVFibro), respectively. At a low serum concentration (10%), which refers to complement concentrations found on mucosal surfaces, the virus was relatively stable at 37 °C. At higher complement levels (up to 50% serum concentration), ZIKV titers differed significantly depending on the cell line used for the propagation of the virus. While the viral titer of ZIKVInsect decreased about two orders in magnitude, when incubated with human serum, the virus derived from human cells was more resistant to complement-mediated lysis (CML). By virus-capture assay and Western blots, the complement regulator protein CD55 was identified to be incorporated into the viral envelope. Blocking of CD55 by neutralizing Abs significantly increased the sensitivity to human complement. Taken together, these data indicate that the incorporation of CD55 from human cells contributes to the stability of ZIKV against complement-mediated virolysis.
Collapse
Affiliation(s)
- Zahra Malekshahi
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Sarah Bernklau
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Britta Schiela
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Iris Koske
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Zoltan Banki
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Claire L. Harris
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Reinhard Würzner
- Institute of Hygiene & Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (R.W.); (H.S.)
| | - Heribert Stoiber
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (Z.M.); (S.B.); (B.S.); (I.K.); (Z.B.)
- Correspondence: (R.W.); (H.S.)
| |
Collapse
|
34
|
Mammalian GPI-anchor modifications and the enzymes involved. Biochem Soc Trans 2021; 48:1129-1138. [PMID: 32573677 DOI: 10.1042/bst20191142] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/26/2022]
Abstract
Glycosylphosphatidylinositol (GPI) is a glycolipid added to the C-terminus of a large variety of proteins in eukaryotes, thereby anchoring these proteins to the cell surface. More than 150 different human proteins are modified with GPI, and GPI-anchored proteins (GPI-APs) play critical roles in embryogenesis, neurogenesis, immunity, and fertilization. GPI-APs are biosynthesized in the endoplasmic reticulum (ER) and transported to the plasma membrane via the Golgi apparatus. During transport, GPI-APs undergo structural remodeling that is important for the efficient folding and sorting of GPI-APs. Asparagine-linked glycan-dependent folding and deacylation by PGAP1 work together to ensure that correctly folded GPI-APs are transported from the ER to the Golgi. Remodeling of the GPI lipid moiety is critical for the association of GPI-APs with lipid rafts. On the cell surface, certain GPI-APs are cleaved by GPI cleavage enzymes and released from the membrane, a key event in processes such as spermatogenesis and neurogenesis. In this review, we discuss the enzymes involved in GPI-AP biosynthesis and the fate of GPI-APs in mammalian cells, with a focus on the assembly, folding, degradation, and cleavage of GPI-APs.
Collapse
|
35
|
Deffieu MS, Cesonyte I, Delalande F, Boncompain G, Dorobantu C, Song E, Lucansky V, Hirschler A, Cianferani S, Perez F, Carapito C, Gaudin R. Rab7-harboring vesicles are carriers of the transferrin receptor through the biosynthetic secretory pathway. SCIENCE ADVANCES 2021; 7:7/2/eaba7803. [PMID: 33523982 PMCID: PMC7793588 DOI: 10.1126/sciadv.aba7803] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 11/18/2020] [Indexed: 06/07/2023]
Abstract
The biosynthetic secretory pathway is particularly challenging to investigate as it is underrepresented compared to the abundance of the other intracellular trafficking routes. Here, we combined the retention using selective hook (RUSH) to a CRISPR-Cas9 gene editing approach (eRUSH) and identified Rab7-harboring vesicles as an important intermediate compartment of the Golgi-to-plasma membrane transport of neosynthesized transferrin receptor (TfR). These vesicles did not exhibit degradative properties and were not associated to Rab6A-harboring vesicles. Rab7A was transiently associated to neosynthetic TfR-containing post-Golgi vesicles but dissociated before fusion with the plasma membrane. Together, our study reveals a role for Rab7 in the biosynthetic secretory pathway of the TfR, highlighting the diversity of the secretory vesicles' nature.
Collapse
Affiliation(s)
- Maika S Deffieu
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS, Univ Montpellier, 34293 Montpellier, France.
- INSERM, Univ Strasbourg, 67000 Strasbourg, France
| | | | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087 Strasbourg, France
| | - Gaelle Boncompain
- Institut Curie, PSL Research University, Sorbonne Université, Centre National de la Recherche Scientifique, UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | | | - Eli Song
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087 Strasbourg, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087 Strasbourg, France
| | - Franck Perez
- Institut Curie, PSL Research University, Sorbonne Université, Centre National de la Recherche Scientifique, UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087 Strasbourg, France
| | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS, Univ Montpellier, 34293 Montpellier, France.
- INSERM, Univ Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
36
|
The Cellular Prion Protein: A Promising Therapeutic Target for Cancer. Int J Mol Sci 2020; 21:ijms21239208. [PMID: 33276687 PMCID: PMC7730109 DOI: 10.3390/ijms21239208] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
Studies on the cellular prion protein (PrPC) have been actively conducted because misfolded PrPC is known to cause transmissible spongiform encephalopathies or prion disease. PrPC is a glycophosphatidylinositol-anchored cell surface glycoprotein that has been reported to affect several cellular functions such as stress protection, cellular differentiation, mitochondrial homeostasis, circadian rhythm, myelin homeostasis, and immune modulation. Recently, it has also been reported that PrPC mediates tumor progression by enhancing the proliferation, metastasis, and drug resistance of cancer cells. In addition, PrPC regulates cancer stem cell properties by interacting with cancer stem cell marker proteins. In this review, we summarize how PrPC promotes tumor progression in terms of proliferation, metastasis, drug resistance, and cancer stem cell properties. In addition, we discuss strategies to treat tumors by modulating the function and expression of PrPC via the regulation of HSPA1L/HIF-1α expression and using an anti-prion antibody.
Collapse
|
37
|
Wang HY, Bharti D, Levental I. Membrane Heterogeneity Beyond the Plasma Membrane. Front Cell Dev Biol 2020; 8:580814. [PMID: 33330457 PMCID: PMC7710808 DOI: 10.3389/fcell.2020.580814] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/02/2020] [Indexed: 01/21/2023] Open
Abstract
The structure and organization of cellular membranes have received intense interest, particularly in investigations of the raft hypothesis. The vast majority of these investigations have focused on the plasma membrane of mammalian cells, yielding significant progress in understanding membrane heterogeneity in terms of lipid composition, molecular structure, dynamic regulation, and functional relevance. In contrast, investigations on lipid organization in other membrane systems have been comparatively scarce, despite the likely relevance of membrane domains in these contexts. In this review, we summarize recent observations on lipid organization in organellar membranes, including endoplasmic reticulum, Golgi, endo-lysosomes, lipid droplets, and secreted membranes like lung surfactant, milk fat globule membranes, and viral membranes. Across these non-plasma membrane systems, it seems that the biophysical principles underlying lipid self-organization contribute to lateral domains.
Collapse
Affiliation(s)
- Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
| | - Deepti Bharti
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
- National Institute of Technology, Rourkela, India
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
38
|
Zhang X, Hao H, Zhuang H, Wang J, Sheng Y, Xu F, Dou J, Chen C, Shen Y. Circular RNA circ_0008305 aggravates hepatocellular carcinoma growth through binding to miR‐186 and inducing TMED2. J Cell Mol Med 2020; 26:1742-1753. [PMID: 33210454 PMCID: PMC8918415 DOI: 10.1111/jcmm.15945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of circRNAs is reported to exert crucial roles in cancers, including hepatocellular carcinoma (HCC). So far, the function of circRNAs in HCC development remains poorly known. Currently, our data showed that circ_0008305 was highly elevated in HCC cell lines and 30 paired tissue samples of HCC. As evidenced, suppression of circ_0008305 repressed HCC cell growth significantly. Meanwhile, up‐regulation of circ_0008305 significantly reduced HCC cell growth. Mechanistically, we displayed that circ_0008305 could bind with miR‐186 by using bioinformatics analysis. miR‐186 has been reported to be a crucial tumour oncogene in many cancers. In addition, we proved miR‐186 was greatly decreased in HCC. The direct correlation between miR‐186 and circ_0008305 was confirmed in our work. In addition, up‐regulation of miR‐186 obviously restrained HCC progression. Increased expression of transmembrane p24 trafficking protein 2 (TMED2) is significantly related to the unfavourable outcomes in cancer patients. At our present work, we proved that TMED2 could act as a direct target of miR‐186. Mechanistically, we demonstrated that circ_0008305 up‐regulated TMED2 expression by sponging miR‐186, which resulted in significantly induced HCC progression in vitro and in vivo. These revealed the significant role of circ_0008305 in HCC progression, which might indicate a new perspective on circRNAs in HCC development.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Hui‐Hui Hao
- Department of Pharmacology Jiangsu College of Nursing Huai'an China
| | - Hai‐Wen Zhuang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Jian Wang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Yu Sheng
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Fang Xu
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Jin Dou
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Chuang Chen
- Division of Hepatobiliary Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Yang Shen
- Operating Room Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| |
Collapse
|
39
|
Jain Goyal M, Zhao X, Bozhinova M, Andrade-López K, de Heus C, Schulze-Dramac S, Müller-McNicoll M, Klumperman J, Béthune J. A paralog-specific role of COPI vesicles in the neuronal differentiation of mouse pluripotent cells. Life Sci Alliance 2020; 3:3/9/e202000714. [PMID: 32665377 PMCID: PMC7368096 DOI: 10.26508/lsa.202000714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 12/03/2022] Open
Abstract
The paralogous COPI coat subunit γ1-COP plays a unique role in promoting neurite outgrowth during the neuronal differentiation of mouse pluripotent cells. Coat protein complex I (COPI)–coated vesicles mediate membrane trafficking between Golgi cisternae as well as retrieval of proteins from the Golgi to the endoplasmic reticulum. There are several flavors of the COPI coat defined by paralogous subunits of the protein complex coatomer. However, whether paralogous COPI proteins have specific functions is currently unknown. Here, we show that the paralogous coatomer subunits γ1-COP and γ2-COP are differentially expressed during the neuronal differentiation of mouse pluripotent cells. Moreover, through a combination of genome editing experiments, we demonstrate that whereas γ-COP paralogs are largely functionally redundant, γ1-COP specifically promotes neurite outgrowth. Our work stresses a role of the COPI pathway in neuronal polarization and provides evidence for distinct functions for coatomer paralogous subunits in this process.
Collapse
Affiliation(s)
- Manu Jain Goyal
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Xiyan Zhao
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Mariya Bozhinova
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Karla Andrade-López
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany.,Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Cecilia de Heus
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sandra Schulze-Dramac
- RNA Regulation Group, Cluster of Excellence "Macromolecular Complexes," Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Michaela Müller-McNicoll
- RNA Regulation Group, Cluster of Excellence "Macromolecular Complexes," Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Julien Béthune
- Junior Research Group, Cluster of Excellence CellNetworks, Heidelberg, Germany .,Heidelberg University Biochemistry Center, Heidelberg, Germany
| |
Collapse
|
40
|
Zhao J, Zhai X, Ma Y, Zhang T, Wang Z, Chong T. Anatomic characteristics of epididymis based on histology, proteomic, and 3D reconstruction. Andrology 2020; 8:1787-1794. [PMID: 32558146 DOI: 10.1111/andr.12842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/20/2020] [Accepted: 06/09/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND The epididymis is a popular research topic in urology and reproduction. OBJECTIVES To explore and identify the anatomical characteristics of the epididymis based on histology, proteomics, and 3D reconstruction of epididymal tubules. MATERIALS AND METHODS A 3D reconstruction of epididymal tubules was generated based on 7-μm-thick transverse serial sections of an epididymis. The proteins in the subcompartments of the epididymis were obtained and analyzed by non-labeled sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH MS). Protein function, signaling pathways, protein expression, and the histology in different subcompartments were analyzed. RESULTS The caput (Cap), corpus (Cor), and cauda (Cau) of the epididymis were divided into 6, 10, and 4 subcompartments, respectively, and the subcompartment between the Cap and Cor is mixed together. A total of 3411 proteins were identified, and 854 proteins were accurately quantified after screening. When the subcompartment Cap 5 transitioned to Cap 6 and Cap 6 to Cap 7, 87 and 52 proteins were upregulated and 14 and 7 proteins were downregulated, respectively. The Cor 9 transition to Cau 1 was marked by 230 proteins that were downregulated, while 74 proteins were upregulated. At the junction of the cauda and the vas deferens, 57 proteins were downregulated, and 410 proteins were upregulated. Cap 6 histology was consistent with that of Cor 1. DISCUSSION AND CONCLUSION The epididymis contains distinct connective tissue septa that can be identified under a surgical tabletop microscope, enabling it to be divided into 20 subcompartments.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| | - Xiaoqiang Zhai
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| | - Yubo Ma
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| | - Tongdian Zhang
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| | - Ziming Wang
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, China
| |
Collapse
|
41
|
Lopez S, Perez-Linero AM, Manzano-Lopez J, Sabido-Bozo S, Cortes-Gomez A, Rodriguez-Gallardo S, Aguilera-Romero A, Goder V, Muñiz M. Dual Independent Roles of the p24 Complex in Selectivity of Secretory Cargo Export from the Endoplasmic Reticulum. Cells 2020; 9:cells9051295. [PMID: 32456004 PMCID: PMC7291304 DOI: 10.3390/cells9051295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 11/22/2022] Open
Abstract
The cellular mechanisms that ensure the selectivity and fidelity of secretory cargo protein transport from the endoplasmic reticulum (ER) to the Golgi are still not well understood. The p24 protein complex acts as a specific cargo receptor for GPI-anchored proteins by facilitating their ER exit through a specialized export pathway in yeast. In parallel, the p24 complex can also exit the ER using the general pathway that exports the rest of secretory proteins with their respective cargo receptors. Here, we show biochemically that the p24 complex associates at the ER with other cargo receptors in a COPII-dependent manner, forming high-molecular weight multireceptor complexes. Furthermore, live cell imaging analysis reveals that the p24 complex is required to retain in the ER secretory cargos when their specific receptors are absent. This requirement does not involve neither the unfolded protein response nor the retrograde transport from the Golgi. Our results suggest that, in addition to its role as a cargo receptor in the specialized GPI-anchored protein pathway, the p24 complex also plays an independent role in secretory cargo selectivity during its exit through the general ER export pathway, preventing the non-selective bulk flow of native secretory cargos. This mechanism would ensure receptor-regulated cargo transport, providing an additional layer of regulation of secretory cargo selectivity during ER export.
Collapse
Affiliation(s)
- Sergio Lopez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Ana Maria Perez-Linero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
| | - Javier Manzano-Lopez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
| | - Susana Sabido-Bozo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Alejandro Cortes-Gomez
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Sofia Rodriguez-Gallardo
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Auxiliadora Aguilera-Romero
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
| | - Veit Goder
- Department of Genetics, University of Seville, 41012 Seville, Spain;
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; (S.L.); (A.M.P.-L.); (J.M.-L.); (S.S.-B.); (A.C.-G.); (S.R.-G.); (A.A.-R.)
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41012 Seville, Spain
- Correspondence: ; Tel.: +34-954556529
| |
Collapse
|
42
|
Kiatpakdee B, Sato K, Otsuka Y, Arashiki N, Chen Y, Tsumita T, Otsu W, Yamamoto A, Kawata R, Yamazaki J, Sugimoto Y, Takada K, Mohandas N, Inaba M. Cholesterol-binding protein TSPO2 coordinates maturation and proliferation of terminally differentiating erythroblasts. J Biol Chem 2020; 295:8048-8063. [PMID: 32358067 PMCID: PMC7278357 DOI: 10.1074/jbc.ra119.011679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/20/2020] [Indexed: 02/02/2023] Open
Abstract
TSPO2 (translocator protein 2) is a transmembrane protein specifically expressed in late erythroblasts and has been postulated to mediate intracellular redistribution of cholesterol. We identified TSPO2 as the causative gene for the HK (high-K+) trait with immature red cell phenotypes in dogs and investigated the effects of the TSPO2 defects on erythropoiesis in HK dogs with the TSPO2 mutation and Tspo2 knockout (Tspo2−/−) mouse models. Bone marrow–derived erythroblasts from HK dogs showed increased binucleated and apoptotic cells at various stages of maturation and shed large nuclei with incomplete condensation when cultured in the presence of erythropoietin, indicating impaired maturation and cytokinesis. The canine TSPO2 induces cholesterol accumulation in the endoplasmic reticulum and could thereby regulate cholesterol availability by changing intracellular cholesterol distribution in erythroblasts. Tspo2−/− mice consistently showed impaired cytokinesis with increased binucleated erythroblasts, resulting in compensated anemia, and their red cell membranes had increased Na,K-ATPase, resembling the HK phenotype in dogs. Tspo2-deficient mouse embryonic stem cell–derived erythroid progenitor (MEDEP) cells exhibited similar morphological defects associated with a cell-cycle arrest at the G2/M phase, resulting in decreased cell proliferation and had a depletion in intracellular unesterified and esterified cholesterol. When the terminal maturation was induced, Tspo2−/− MEDEP cells showed delays in hemoglobinization; maturation-associated phenotypic changes in CD44, CD71, and TER119 expression; and cell-cycle progression. Taken together, these findings imply that TSPO2 is essential for coordination of maturation and proliferation of erythroblasts during normal erythropoiesis.
Collapse
Affiliation(s)
- Benjaporn Kiatpakdee
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kota Sato
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yayoi Otsuka
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuto Arashiki
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuqi Chen
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Tsumita
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Wataru Otsu
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Akito Yamamoto
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Reo Kawata
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Jumpei Yamazaki
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | - Kensuke Takada
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
43
|
Abstract
At least 150 human proteins are glycosylphosphatidylinositol-anchored proteins (GPI-APs). The protein moiety of GPI-APs lacking transmembrane domains is anchored to the plasma membrane with GPI covalently attached to the C-terminus. The GPI consists of the conserved core glycan, phosphatidylinositol and glycan side chains. The entire GPI-AP is anchored to the outer leaflet of the lipid bilayer by insertion of fatty chains of phosphatidylinositol. Because of GPI-dependent membrane anchoring, GPI-APs have some unique characteristics. The most prominent feature of GPI-APs is their association with membrane microdomains or membrane rafts. In the polarized cells such as epithelial cells, many GPI-APs are exclusively expressed in the apical surfaces, whereas some GPI-APs are preferentially expressed in the basolateral surfaces. Several GPI-APs act as transcytotic transporters carrying their ligands from one compartment to another. Some GPI-APs are shed from the membrane after cleavage within the GPI by a GPI-specific phospholipase or a glycosidase. In this review, I will summarize the current understanding of GPI-AP biosynthesis in mammalian cells and discuss examples of GPI-dependent functions of mammalian GPI-APs.
Collapse
Affiliation(s)
- Taroh Kinoshita
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
44
|
Abstract
Regulated transport through the secretory pathway is essential for embryonic development and homeostasis. Disruptions in this process impact cell fate, differentiation and survival, often resulting in abnormalities in morphogenesis and in disease. Several congenital malformations are caused by mutations in genes coding for proteins that regulate cargo protein transport in the secretory pathway. The severity of mutant phenotypes and the unclear aetiology of transport protein-associated pathologies have motivated research on the regulation and mechanisms through which these proteins contribute to morphogenesis. This review focuses on the role of the p24/transmembrane emp24 domain (TMED) family of cargo receptors in development and disease.
Collapse
|
45
|
Endoplasmic Reticulum Export of GPI-Anchored Proteins. Int J Mol Sci 2019; 20:ijms20143506. [PMID: 31319476 PMCID: PMC6678536 DOI: 10.3390/ijms20143506] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/07/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Protein export from the endoplasmic reticulum (ER) is an essential process in all eukaryotes driven by the cytosolic coat complex COPII, which forms vesicles at ER exit sites for transport of correctly assembled secretory cargo to the Golgi apparatus. The COPII machinery must adapt to the existing wide variety of different types of cargo proteins and to different cellular needs for cargo secretion. The study of the ER export of glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs), a special glycolipid-linked class of cell surface proteins, is contributing to address these key issues. Due to their special biophysical properties, GPI-APs use a specialized COPII machinery to be exported from the ER and their processing and maturation has been recently shown to actively regulate COPII function. In this review, we discuss the regulatory mechanisms by which GPI-APs are assembled and selectively exported from the ER.
Collapse
|
46
|
Funato K, Riezman H, Muñiz M. Vesicular and non-vesicular lipid export from the ER to the secretory pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158453. [PMID: 31054928 DOI: 10.1016/j.bbalip.2019.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 11/26/2022]
Abstract
The endoplasmic reticulum is the site of synthesis of most glycerophospholipids, neutral lipids and the initial steps of sphingolipid biosynthesis of the secretory pathway. After synthesis, these lipids are distributed within the cells to create and maintain the specific compositions of the other secretory organelles. This represents a formidable challenge, particularly while there is a simultaneous and quantitatively important flux of membrane components stemming from the vesicular traffic of proteins through the pathway, which can also vary depending on the cell type and status. To meet this challenge cells have developed an intricate system of interorganellar contacts and lipid transport proteins, functioning in non-vesicular lipid transport, which are able to ensure membrane lipid homeostasis even in the absence of membrane trafficking. Nevertheless, under normal conditions, lipids are transported in cells by both vesicular and non-vesicular mechanisms. In this review we will discuss the mechanism and roles of vesicular and non-vesicular transport of lipids from the ER to other organelles of the secretory pathway.
Collapse
Affiliation(s)
- Kouichi Funato
- Department of Bioresource Science and Technology, Hiroshima University, Japan.
| | - Howard Riezman
- NCCR Chemical Biology and Department of Biochemistry, Sciences II, University of Geneva, Switzerland.
| | - Manuel Muñiz
- Department of Cell Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| |
Collapse
|
47
|
Zhang X, Wu Y, Cai F, Song W. Regulation of global gene expression in brain by TMP21. Mol Brain 2019; 12:39. [PMID: 31036051 PMCID: PMC6489340 DOI: 10.1186/s13041-019-0460-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
TMP21, a type I transmembrane protein of thep24 protein family, mediates protein trafficking and maturation. Dysregulation of TMP21 is implicated in the pathogenesis of Alzheimer’s disease (AD). However, underlying mechanisms remain elusive. To reveal the function of TMP21 in the brain and the pathogenic role of TMP21 in the brain of AD, the global gene expression was profiled in the brain of TMP21 knockdown mice. We found that 8196 and 8195 genes are significantly altered in the hippocampus and cortex, respectively. The genes are involved in a number of brain function-related pathways, including glutamatergic synapse pathway, serotonergic synapse pathway, synaptic vesicle pathway, and long-term depression pathway. Moreover, the network analysis suggests that the TMP21 may contribute to the pathogenesis of AD by regulatingPI3K/Akt/GSK3β signalling pathway. Our study provides an insight into the physiological function of TMP21 in the brain and pathological role of TMP21 in AD.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Mental Disorders, Changsha, Hunan, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong, China
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
48
|
Abstract
The Golgi apparatus is a central intracellular membrane-bound organelle with key functions in trafficking, processing, and sorting of newly synthesized membrane and secretory proteins and lipids. To best perform these functions, Golgi membranes form a unique stacked structure. The Golgi structure is dynamic but tightly regulated; it undergoes rapid disassembly and reassembly during the cell cycle of mammalian cells and is disrupted under certain stress and pathological conditions. In the past decade, significant amount of effort has been made to reveal the molecular mechanisms that regulate the Golgi membrane architecture and function. Here we review the major discoveries in the mechanisms of Golgi structure formation, regulation, and alteration in relation to its functions in physiological and pathological conditions to further our understanding of Golgi structure and function in health and diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Adolf F, Rhiel M, Hessling B, Gao Q, Hellwig A, Béthune J, Wieland FT. Proteomic Profiling of Mammalian COPII and COPI Vesicles. Cell Rep 2019; 26:250-265.e5. [DOI: 10.1016/j.celrep.2018.12.041] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 09/08/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
|
50
|
Kovalchuk V, Samluk Ł, Juraszek B, Jurkiewicz-Trząska D, Sucic S, Freissmuth M, Nałęcz KA. Trafficking of the amino acid transporter B 0,+ (SLC6A14) to the plasma membrane involves an exclusive interaction with SEC24C for its exit from the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:252-263. [PMID: 30445147 PMCID: PMC6314439 DOI: 10.1016/j.bbamcr.2018.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 11/02/2018] [Accepted: 11/12/2018] [Indexed: 12/16/2022]
Abstract
A plasma membrane amino acid transporter B0,+ (ATB0,+), encoded by the SLC6A14 gene, is specific for neutral and basic amino acids. It is up-regulated in several types of malignant cancers. Neurotransmitter transporters of the SLC6 family interact with specific SEC24 proteins of the COPII complex along their pathway from the endoplasmic reticulum (ER) to Golgi. This study focused on the possible role of SEC24 proteins in ATB0,+ trafficking. Rat ATB0,+ was expressed in HEK293 cells, its localization and trafficking were examined by Western blot, deglycosylation, immunofluorescence (co-localization with ER and trans-Golgi markers) and biotinylation. The expression of ATB0,+ at the plasma membrane was decreased by dominant negative mutants of SAR1, a GTPase, whose activity triggers the formation of the COPII complex. ATB0,+ co-precipitated with SEC24C (but not with the remaining isoforms A, B and D). This interaction was confirmed by immunocytochemistry and the proximity ligation assay. Co-localization of SEC24C with endogenous ATB0,+ was also observed in MCF-7 breast cancer cells. Contrary to the endogenous transporter, part of the overexpressed ATB0,+ is directed to proteolysis, a process significantly reversed by a proteasome inhibitor bortezomib. Co-transfection with a SEC24C dominant negative mutant attenuated ATB0,+ expression at the plasma membrane, due to proteolytic degradation. These results support a hypothesis that lysine at position +2 downstream of the ER export "RI" motif on the cargo protein is crucial for SEC24C binding and for further trafficking to the Golgi. Moreover, there is an equilibrium between ER export and degradation mechanisms in case of overexpressed transporter.
Collapse
Affiliation(s)
- Vasylyna Kovalchuk
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Łukasz Samluk
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Barbara Juraszek
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Dominika Jurkiewicz-Trząska
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Katarzyna A Nałęcz
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|