1
|
Huang KY, Yu HC, Lu MC, Tseng HYH, Shen JJ, Lin CY, Chen PC, Shen YT, Chung PR, Tsai HK, Zhou SR, Wang CL, Lai NS, Lin TH, Huang HB. Identification of a novel Eps 15 homology domain-containing protein 1 (EHD1) and EHD4-binding motif in phostensin. J Biochem 2025; 177:297-304. [PMID: 39776131 DOI: 10.1093/jb/mvaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Phostensin (PTS) encoded by KIAA1949 binds to protein phosphatase 1, F-actin, Eps 15 homology domain-containing protein 1 (EHD1) and EHD4. Most EHD-binding proteins contain a consensus motif, Asn-Pro-Phe (NPF), which interacts with the C-terminal EH domain of EHD proteins. Nevertheless, the NPF motif is absent in PTS. The binding motif for PTS to interact with EHD1 (or EHD4) remains unknown. Here, we identified that PTS-α binds to EHD1 (or EHD4) through the region of residues 51-80, which contains a consensus motif, 64ILV(X)4(L/V)RL74S. This novel consensus motif is also found in vacuolar protein sorting-35 (vps35). Replacement of 64ILV(X)4(L/V)RL74S with 64AAA(X)4(L/V)RL74S or with 64ILV(X)4AEA74A significantly reduces the binding efficiency of PTS-α to either EHD1 or EHD4 in GST pull-down assay and far western blotting assay. In addition, replacement of 218ILV(X)4VRL228S with 218AAA(X)4AEA228A decreases the binding ability of vps35 to EHD4 in far western blotting assay. Overexpression of the PTS-β in 293 T cells attenuated the endocytic trafficking of transferrin. However, this attenuation of transferrin in endocytic trafficking was disrupted when 293 T cells overexpressed the mutant PTS-β with a defective EHD-binding motif, suggesting that PTS-β can regulate the endocytic recycling via associating with EHD1 or EHD4.
Collapse
Affiliation(s)
- Kuang-Yung Huang
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
| | - Hui-Chun Yu
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Hsien-Yu Huang Tseng
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Jyun-Jie Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Ying Lin
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pin-Chen Chen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Ya-Ting Shen
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Pei-Rong Chung
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Hsiao-Kuei Tsai
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Si-Ru Zhou
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| | - Chia-Lin Wang
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Ning-Sheng Lai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
- School of Medicine, Tzu Chi University, Sec. 3, Zhongyang Rd., Hualien 970, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Min-Sheng Rd., Chia-Yi 62247, Taiwan
| | - Ta-Hsien Lin
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Sec. 2, Linong St., Taipei 11221, Taiwan
| | - Hsien-Bin Huang
- Department of Biomedical Sciences, National Chung Cheng University, Sec. 1, University Rd., Chia-Yi 621, Taiwan
| |
Collapse
|
2
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of Caenorhabditis elegans. Genetics 2025; 229:iyae216. [PMID: 39722491 DOI: 10.1093/genetics/iyae216] [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: 09/15/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Membrane trafficking is a conserved process required for the import, export, movement, and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. Using a genetic approach, we identified reduction-of-function mutations in tat-1 that suppress nekl-associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine flippase that promotes the asymmetric distribution of phosphatidylserine on the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a phosphatidylserine sensor, we found that TAT-1 was required for the normal localization of phosphatidylserine at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with these data, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology domain-containing protein, EHD1. TAT-1, phosphatidylserine biosynthesis, and the phosphatidylserine-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did inhibition of phosphatidylserine biosynthesis. We propose that TAT-1 flippase activity, in conjunction with RFIP-2, promotes the recruitment of RME-1 to apical recycling endosomes and that inhibition of TAT-1-RFIP-2-RME-1 can compensate for a reduction in NEKL activities.
Collapse
Affiliation(s)
- Shae M Milne
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - Philip T Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - David S Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| |
Collapse
|
3
|
Fowler H, Clifford RE, Bowden D, Sutton PA, Govindarajah N, Fok M, Glenn M, Wall M, Rubbi C, Buczacki SJA, Mandal A, Francies H, Hughes J, Parsons JL, Vimalachandran D. Myoferlin: A Potential Marker of Response to Radiation Therapy and Survival in Locally Advanced Rectal Cancer. Int J Radiat Oncol Biol Phys 2024; 120:1111-1123. [PMID: 38866213 DOI: 10.1016/j.ijrobp.2024.05.030] [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: 08/03/2023] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Patients with locally advanced rectal cancer often require neoadjuvant chemoradiation therapy to downstage the disease, but the response is variable with no predictive biomarkers. We have previously revealed through proteomic profiling that myoferlin is associated with response to radiation therapy. The aims of this study were to further validate this finding and explore the potential for myoferlin to act as a prognostic and/or therapeutic target. METHODS AND MATERIALS Immunohistochemical analysis of a tissue microarray (TMA) for 111 patients was used to validate the initial proteomic findings. Manipulation of myoferlin was achieved using small interfering RNA, a small molecular inhibitor (wj460), and a CRISPR-Cas9 knockout cell line. Radiosensitization after treatment was assessed using 2-dimensional clonogenic assays, 3-dimensional spheroid models, and patient-derived organoids. Underlying mechanisms were investigated using electrophoresis, immunofluorescence, and immunoblotting. RESULTS Analysis of both the diagnostic biopsy and tumor resection samples confirmed that low myoferlin expression correlated with a good response to neoadjuvant long-course chemoradiation therapy. High myoferlin expression was associated with spread to local lymph nodes and worse 5-year survival (P = .01; hazard ratio, 3.5; 95% CI, 1.27-10.04). This was externally validated using the Stratification in Colorectal Cancer database. Quantification of myoferlin using immunoblotting in immortalized colorectal cancer cell lines and organoids demonstrated that high myoferlin expression was associated with increased radioresistance. Biological and pharmacologic manipulation of myoferlin resulted in significantly increased radiosensitivity across all cell lines in 2-dimensional and 3-dimensional models. After irradiation, myoferlin knockdown cells had a significantly impaired ability to repair DNA double-strand breaks. This appeared to be mediated via nonhomologous end-joining. CONCLUSIONS We have confirmed that high expression of myoferlin in rectal cancer is associated with poor response to neoadjuvant therapy and worse long-term survival. Furthermore, the manipulation of myoferlin led to increased radiosensitivity in vitro. This suggests that myoferlin could be targeted to enhance the sensitivity of patients with rectal cancer to radiation therapy, and further work is required.
Collapse
Affiliation(s)
- Hayley Fowler
- Department of Colorectal Surgery, Countess of Chester National Health Service Foundation Trust, Chester, United Kingdom; Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom.
| | - Rachael E Clifford
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| | - David Bowden
- Department of Colorectal Surgery, Countess of Chester National Health Service Foundation Trust, Chester, United Kingdom
| | - Paul A Sutton
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| | - Naren Govindarajah
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| | - Matthew Fok
- Department of Colorectal Surgery, Countess of Chester National Health Service Foundation Trust, Chester, United Kingdom; Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| | - Mark Glenn
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| | - Michael Wall
- Department of Colorectal Surgery, Countess of Chester National Health Service Foundation Trust, Chester, United Kingdom
| | - Carlos Rubbi
- Faculty of Health, Social Care & Medicine, Edge Hill University, St Helens Road, Ormskirk, Lancashire, L39 4QP
| | - Simon J A Buczacki
- Nuffield Department of Surgical Sciences (NDS), University of Oxford, Oxford, United Kingdom
| | - Amit Mandal
- Nuffield Department of Surgical Sciences (NDS), University of Oxford, Oxford, United Kingdom
| | - Hayley Francies
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan Hughes
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jason L Parsons
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dale Vimalachandran
- Department of Colorectal Surgery, Countess of Chester National Health Service Foundation Trust, Chester, United Kingdom; Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, William Henry Duncan Building, Liverpool, United Kingdom
| |
Collapse
|
4
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613099. [PMID: 39314363 PMCID: PMC11419146 DOI: 10.1101/2024.09.15.613099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Membrane trafficking is a conserved process required for the movement and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. We used a genetic approach to identify reduction-of-function mutations in tat-1 that suppress nekl -associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine (PS) flippase that promotes the asymmetric distribution of PS to the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a PS sensor, we found that TAT-1 was required for the normal localization of PS at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with this, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology (EH) domain-containing protein, EHD1. TAT-1, PS biosynthesis, and the PS-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did the inhibition of PS biosynthesis. Using the auxin-inducible degron system, we found that depletion of NEKL-2 or NEKL-3 led to defects in RME-1 localization and that a reduction in TAT-1 function partially restored RME-1 localization in NEKL-3-depleted cells. ARTICLE SUMMARY Endocytosis is an essential process required for the movement of proteins and lipids within cells. NEKL-2 and NEKL-3, two evolutionarily conserved proteins in the nematode Caenorhabditis elegans , are important regulators of endocytosis. In the current study, the authors describe a new functional link between the NEKLs and several proteins with known roles in endocytosis including TAT-1, a conserved enzyme that moves lipids between the bilayers of cellular membranes. As previous work implicated NEKLs in developmental defects and cancer, the present study can provide new insights into how the misregulation of endocytosis affects human health and disease.
Collapse
|
5
|
Harders RH, Morthorst TH, Landgrebe LE, Lande AD, Fuglsang MS, Mortensen SB, Feteira-Montero V, Jensen HH, Wesseltoft JB, Olsen A. CED-6/GULP and components of the clathrin-mediated endocytosis machinery act redundantly to correctly display CED-1 on the cell membrane in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2024; 14:jkae088. [PMID: 38696649 PMCID: PMC11228867 DOI: 10.1093/g3journal/jkae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/04/2024]
Abstract
CED-1 (cell death abnormal) is a transmembrane receptor involved in the recognition of "eat-me" signals displayed on the surface of apoptotic cells and thus central for the subsequent engulfment of the cell corpse in Caenorhabditis elegans. The roles of CED-1 in engulfment are well established, as are its downstream effectors. The latter include the adapter protein CED-6/GULP and the ATP-binding cassette family homolog CED-7. However, how CED-1 is maintained on the plasma membrane in the absence of engulfment is currently unknown. Here, we show that CED-6 and CED-7 have a novel role in maintaining CED-1 correctly on the plasma membrane. We propose that the underlying mechanism is via endocytosis as CED-6 and CED-7 act redundantly with clathrin and its adaptor, the Adaptor protein 2 complex, in ensuring correct CED-1 localization. In conclusion, CED-6 and CED-7 impact other cellular processes than engulfment of apoptotic cells.
Collapse
Affiliation(s)
- Rikke Hindsgaul Harders
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Tine H Morthorst
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus, DK-8000, Denmark
| | - Line E Landgrebe
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus, DK-8000, Denmark
| | - Anna D Lande
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus, DK-8000, Denmark
| | - Marie Sikjær Fuglsang
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus, DK-8000, Denmark
| | - Stine Bothilde Mortensen
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Verónica Feteira-Montero
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Helene Halkjær Jensen
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Jonas Bruhn Wesseltoft
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Anders Olsen
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| |
Collapse
|
6
|
Jiménez-Guerrero I, Sonawane M, Eckshtain-Levi N, Tuang ZK, da Silva GM, Pérez-Montaño F, Leibman-Markus M, Gupta R, Noda-Garcia L, Bar M, Burdman S. Natural variation in a short region of the Acidovorax citrulli type III-secreted effector AopW1 is associated with differences in cytotoxicity and host adaptation. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 117:516-540. [PMID: 37864805 DOI: 10.1111/tpj.16507] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023]
Abstract
Bacterial fruit blotch, caused by Acidovorax citrulli, is a serious disease of melon and watermelon. The strains of the pathogen belong to two major genetic groups: group I strains are strongly associated with melon, while group II strains are more aggressive on watermelon. A. citrulli secretes many protein effectors to the host cell via the type III secretion system. Here we characterized AopW1, an effector that shares similarity to the actin cytoskeleton-disrupting effector HopW1 of Pseudomonas syringae and with effectors from other plant-pathogenic bacterial species. AopW1 has a highly variable region (HVR) within amino acid positions 147 to 192, showing 14 amino acid differences between group I and II variants. We show that group I AopW1 is more toxic to yeast and Nicotiana benthamiana cells than group II AopW1, having stronger actin filament disruption activity, and increased ability to induce cell death and reduce callose deposition. We further demonstrated the importance of some amino acid positions within the HVR for AopW1 cytotoxicity. Cellular analyses revealed that AopW1 also localizes to the endoplasmic reticulum, chloroplasts, and plant endosomes. We also show that overexpression of the endosome-associated protein EHD1 attenuates AopW1-induced cell death and increases defense responses. Finally, we show that sequence variation in AopW1 plays a significant role in the adaptation of group I and II strains to their preferred hosts, melon and watermelon, respectively. This study provides new insights into the HopW1 family of bacterial effectors and provides first evidence on the involvement of EHD1 in response to biotic stress.
Collapse
Affiliation(s)
- Irene Jiménez-Guerrero
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Monica Sonawane
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Noam Eckshtain-Levi
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Za Khai Tuang
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Gustavo Mateus da Silva
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Francisco Pérez-Montaño
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
- Department of Microbiology, University of Seville, Seville, Spain
| | - Meirav Leibman-Markus
- Department of Plant Pathology and Weed Research, Agricultural Research Organization, The Volcani Institute, Bet Dagan, Israel
| | - Rupali Gupta
- Department of Plant Pathology and Weed Research, Agricultural Research Organization, The Volcani Institute, Bet Dagan, Israel
| | - Lianet Noda-Garcia
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Maya Bar
- Department of Plant Pathology and Weed Research, Agricultural Research Organization, The Volcani Institute, Bet Dagan, Israel
| | - Saul Burdman
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Environmental Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
7
|
Cheng X, Li X, Yang M, Zheng C, Li H, Qu L, Ning Z. Genome-wide association study exploring the genetic architecture of eggshell speckles in laying hens. BMC Genomics 2023; 24:704. [PMID: 37993775 PMCID: PMC10666442 DOI: 10.1186/s12864-023-09632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/28/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Eggshell speckle phenotype is an important trait in poultry production because they affect eggshell quality. However, the genetic architecture of speckled eggshells remains unclear. In this study, we determined the heritability of eggshell speckles and conducted a genome-wide association study (GWAS) on purebred Rhode Island Red (RIR) hens at 28 weeks to detect potential genomic loci and candidate genes associated with eggshell speckles. RESULTS The heritability of eggshell speckles was 0.35 at 28 weeks, and the speckle level is not related to other eggshell quality traits in terms of phenotypic correlation. We detected 311 SNPs (6 significantly, and 305 suggestively associated) and 39 candidate genes associated with eggshell speckles. Based on the pathway analysis, the 39 candidate genes were mainly involved in alpha-linolenic acid metabolism, linoleic acid metabolism, ether lipid metabolism, GnRH signaling pathway, vascular smooth muscle contraction, and MAPK signaling pathway. Ultimately, ten genes, LOC423226, SPTBN5, EHD4, LOC77155, TYRO3, ITPKA, DLL4, PLA2G4B, PLA2G4EL5, and PLA2G4EL6 were considered the most promising genes associated with eggshell speckles that were implicated in immunoregulation, calcium transport, and phospholipid metabolism, while its function in laying hens requires further studies. CONCLUSIONS This study provides new insights into understanding the genetic basis of eggshell speckles and has practical application value for the genetic improvement of eggshell quality.
Collapse
Affiliation(s)
- Xue Cheng
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xinghua Li
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mengyuan Yang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chuanwei Zheng
- Beijing Zhongnongbangyang Layer Breeding Co., Ltd, Beijing, 100083, China
| | - Haiying Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830000, China
| | - Lujiang Qu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Zhonghua Ning
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Finch NC, Neal CR, Welsh GI, Foster RR, Satchell SC. The unique structural and functional characteristics of glomerular endothelial cell fenestrations and their potential as a therapeutic target in kidney disease. Am J Physiol Renal Physiol 2023; 325:F465-F478. [PMID: 37471420 PMCID: PMC10639027 DOI: 10.1152/ajprenal.00036.2023] [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: 02/21/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Glomerular endothelial cell (GEnC) fenestrations are a critical component of the glomerular filtration barrier. Their unique nondiaphragmed structure is key to their function in glomerular hydraulic permeability, and their aberration in disease can contribute to loss of glomerular filtration function. This review provides a comprehensive update of current understanding of the regulation and biogenesis of fenestrae. We consider diseases in which GEnC fenestration loss is recognized or may play a role and discuss methods with potential to facilitate the study of these critical structures. Literature is drawn from GEnCs as well as other fenestrated cell types such as liver sinusoidal endothelial cells that most closely parallel GEnCs.
Collapse
Affiliation(s)
- Natalie C Finch
- Bristol Renal, University of Bristol, United Kingdom
- Langford Vets, University of Bristol, United Kingdom
| | - Chris R Neal
- Bristol Renal, University of Bristol, United Kingdom
| | - Gavin I Welsh
- Bristol Renal, University of Bristol, United Kingdom
| | | | | |
Collapse
|
9
|
Naslavsky N, Caplan S. Advances and challenges in understanding endosomal sorting and fission. FEBS J 2023; 290:4187-4195. [PMID: 36413090 PMCID: PMC10200825 DOI: 10.1111/febs.16687] [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: 09/20/2022] [Revised: 11/04/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
Endosomes play crucial roles in the cell, serving as focal and 'triage' points for internalized lipids and receptors. As such, endosomes are a critical branching point that determines whether receptors are sorted for degradation or recycling. This Viewpoint aims to highlight recent advances in endosome research, including key endosomal functions such as sorting and fission. Moreover, the Viewpoint addresses key technical and conceptual challenges in studying endosomes.
Collapse
Affiliation(s)
- Naava Naslavsky
- Department of Biochemistry & Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
10
|
Yu CW, Wu YC, Liao VHC. Nanoplastics exposure disrupts circadian rhythm associated with dysfunction of the endolysosomal pathway and autophagy in Caenorhabditis elegans. JOURNAL OF HAZARDOUS MATERIALS 2023; 452:131308. [PMID: 37004444 DOI: 10.1016/j.jhazmat.2023.131308] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/15/2023] [Accepted: 03/26/2023] [Indexed: 05/03/2023]
Abstract
Nanoplastics (NPs), an emerging pollutant, have raised great safety concerns due to their widespread applications and continuous release into the environment, which lead to potential human and environmental risks. Recently, polystyrene NPs (100 nm; 100 mg/L) exposure has been reported to disrupt circadian rhythms under five days temperature entrainment and be associated with stress resistance decline in Caenorhabditis elegans. This study explored the possible relationship between circadian rhythm disruption and endocytosis and autophagy under polystyrene NPs exposure in C. elegans. We show that the disrupted circadian rhythm induced by NPs exposure reduced stress resistance via endocytosis and autophagy impairment. Furthermore, we found that most NPs taken up by intestinal cells were localized to early endosomes, late endosomes, and lysosomes and delivered to autophagosomes. In addition, the disruption of circadian rhythm inhibited NPs localization to these organelles. These findings indicate that NPs exposure disrupts circadian rhythm and alters its subcellular trafficking, leading to enhanced toxicity in C. elegans. Our results shed light on the prominent role of NPs exposure in circadian rhythm disruption associated with endocytosis and autophagy impairments, which may be conserved in higher animals such as humans.
Collapse
Affiliation(s)
- Chan-Wei Yu
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, ROC
| | - Yi-Chun Wu
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, ROC
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, ROC.
| |
Collapse
|
11
|
Hernandez-Perez I, Rubio J, Baumann A, Girao H, Ferrando M, Rebollo E, Aragay AM, Geli MI. Kazrin promotes dynein/dynactin-dependent traffic from early to recycling endosomes. eLife 2023; 12:e83793. [PMID: 37096882 PMCID: PMC10181827 DOI: 10.7554/elife.83793] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/24/2023] [Indexed: 04/26/2023] Open
Abstract
Kazrin is a protein widely expressed in vertebrates whose depletion causes a myriad of developmental defects, in part derived from altered cell adhesion and migration, as well as failure to undergo epidermal to mesenchymal transition. However, the primary molecular role of kazrin, which might contribute to all these functions, has not been elucidated yet. We previously identified one of its isoforms, kazrin C, as a protein that potently inhibits clathrin-mediated endocytosis when overexpressed. We now generated kazrin knock-out mouse embryonic fibroblasts to investigate its endocytic function. We found that kazrin depletion delays juxtanuclear enrichment of internalized material, indicating a role in endocytic traffic from early to recycling endosomes. Consistently, we found that the C-terminal domain of kazrin C, predicted to be an intrinsically disordered region, directly interacts with several early endosome (EE) components, and that kazrin depletion impairs retrograde motility of these organelles. Further, we noticed that the N-terminus of kazrin C shares homology with dynein/dynactin adaptors and that it directly interacts with the dynactin complex and the dynein light intermediate chain 1. Altogether, the data indicate that one of the primary kazrin functions is to facilitate endocytic recycling by promoting dynein/dynactin-dependent transport of EEs or EE-derived transport intermediates to the recycling endosomes.
Collapse
Affiliation(s)
- Ines Hernandez-Perez
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Javier Rubio
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Adrian Baumann
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Henrique Girao
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Miriam Ferrando
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Elena Rebollo
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - Anna M Aragay
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| | - María Isabel Geli
- Institute for Molecular Biology of Barcelona (IBMB, CSIC), Baldiri Reixac 15BarcelonaSpain
| |
Collapse
|
12
|
Kustigian L, Gong X, Gai W, Thongchol J, Zhang J, Puchalla J, Carr CM, Rye HS. GTP-stimulated membrane fission by the N-BAR protein AMPH-1. Traffic 2023; 24:34-47. [PMID: 36435193 PMCID: PMC9825645 DOI: 10.1111/tra.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 10/24/2022] [Accepted: 11/19/2022] [Indexed: 11/28/2022]
Abstract
Membrane-enclosed transport carriers sort biological molecules between stations in the cell in a dynamic process that is fundamental to the physiology of eukaryotic organisms. While much is known about the formation and release of carriers from specific intracellular membranes, the mechanism of carrier formation from the recycling endosome, a compartment central to cellular signaling, remains to be resolved. In Caenorhabditis elegans, formation of transport carriers from the recycling endosome requires the dynamin-like, Eps15-homology domain (EHD) protein, RME-1, functioning with the Bin/Amphiphysin/Rvs (N-BAR) domain protein, AMPH-1. Here we show, using a free-solution single-particle technique known as burst analysis spectroscopy (BAS), that AMPH-1 alone creates small, tubular-vesicular products from large, unilamellar vesicles by membrane fission. Membrane fission requires the amphipathic H0 helix of AMPH-1 and is slowed in the presence of RME-1. Unexpectedly, AMPH-1-induced membrane fission is stimulated in the presence of GTP. Furthermore, the GTP-stimulated membrane fission activity seen for AMPH-1 is recapitulated by the heterodimeric N-BAR amphiphysin protein from yeast, Rvs161/167p, strongly suggesting that GTP-stimulated membrane fission is a general property of this important class of N-BAR proteins.
Collapse
Affiliation(s)
- Lauren Kustigian
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
- Current address: GlaxoSmithKline, 1250 South Collegeville Rd., Collegeville, Pennsylvania 19426, USA
| | - Xue Gong
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| | - Wei Gai
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| | - Jirapat Thongchol
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| | - Junjie Zhang
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| | - Jason Puchalla
- Department of Physics, Princeton University, Princeton, New Jersey 08544, USA
| | - Chavela M. Carr
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| | - Hays S. Rye
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, 77845, USA
| |
Collapse
|
13
|
Finch NC, Fawaz SS, Neal CR, Butler MJ, Lee VK, Salmon AJ, Lay AC, Stevens M, Dayalan L, Band H, Mellor HH, Harper SJ, Shima DT, Welsh GI, Foster RR, Satchell SC. Reduced Glomerular Filtration in Diabetes Is Attributable to Loss of Density and Increased Resistance of Glomerular Endothelial Cell Fenestrations. J Am Soc Nephrol 2022; 33:1120-1136. [PMID: 35292439 PMCID: PMC9161794 DOI: 10.1681/asn.2021030294] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/01/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Glomerular endothelial cell (GEnC) fenestrations are recognized as an essential component of the glomerular filtration barrier, yet little is known about how they are regulated and their role in disease. METHODS We comprehensively characterized GEnC fenestral and functional renal filtration changes including measurement of glomerular Kf and GFR in diabetic mice (BTBR ob-/ob- ). We also examined and compared human samples. We evaluated Eps homology domain protein-3 (EHD3) and its association with GEnC fenestrations in diabetes in disease samples and further explored its role as a potential regulator of fenestrations in an in vitro model of fenestration formation using b.End5 cells. RESULTS Loss of GEnC fenestration density was associated with decreased filtration function in diabetic nephropathy. We identified increased diaphragmed fenestrations in diabetes, which are posited to increase resistance to filtration and further contribute to decreased GFR. We identified decreased glomerular EHD3 expression in diabetes, which was significantly correlated with decreased fenestration density. Reduced fenestrations in EHD3 knockdown b.End5 cells in vitro further suggested a mechanistic role for EHD3 in fenestration formation. CONCLUSIONS This study demonstrates the critical role of GEnC fenestrations in renal filtration function and suggests EHD3 may be a key regulator, loss of which may contribute to declining glomerular filtration function through aberrant GEnC fenestration regulation. This points to EHD3 as a novel therapeutic target to restore filtration function in disease.
Collapse
Affiliation(s)
- Natalie C. Finch
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sarah S. Fawaz
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Chris R. Neal
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Matthew J. Butler
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Vivian K. Lee
- Translational Vision Research, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Andrew J. Salmon
- Renal Service, Waitemata District Health Board, Auckland, New Zealand
| | - Abigail C. Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Megan Stevens
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Lusyan Dayalan
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Hamid Band
- Eppley Institute for Research in Cancer, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Harry H. Mellor
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Steven J. Harper
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - David T. Shima
- Translational Vision Research, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Gavin I. Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Rebecca R. Foster
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Simon C. Satchell
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
14
|
Jones T, Naslavsky N, Caplan S. Differential requirements for the Eps15 homology Domain Proteins EHD4 and EHD2 in the regulation of mammalian ciliogenesis. Traffic 2022; 23:360-373. [PMID: 35510564 PMCID: PMC9324998 DOI: 10.1111/tra.12845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022]
Abstract
The endocytic protein EHD1 controls primary ciliogenesis by facilitating fusion of the ciliary vesicle and by removal of CP110 from the mother centriole. EHD3, the closest EHD1 paralog, has a similar regulatory role, but initial evidence suggested that the other two more distal paralogs, EHD2 and EHD4 may be dispensable for ciliogenesis. Herein, we define a novel role for EHD4, but not EHD2, in regulating primary ciliogenesis. To better understand the mechanisms and differential functions of the EHD proteins in ciliogenesis, we first demonstrated a requirement for EHD1 ATP‐binding to promote ciliogenesis. We then identified two sequence motifs that are entirely conserved between EH domains of EHD1, EHD3 and EHD4, but display key amino acid differences within the EHD2 EH domain. Substitution of either P446 or E470 in EHD1 with the aligning S451 or W475 residues from EHD2 was sufficient to prevent rescue of ciliogenesis in EHD1‐depleted cells upon reintroduction of EHD1. Overall, our data enhance the current understanding of the EHD paralogs in ciliogenesis, demonstrate a need for ATP‐binding and identify conserved sequences in the EH domains of EHD1, EHD3 and EHD4 that regulate EHD1 binding to proteins and its ability to rescue ciliogenesis in EHD1‐depleted cells.
Collapse
Affiliation(s)
- Tyler Jones
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE
| | - Naava Naslavsky
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
15
|
Hasegawa J, Uchida Y, Mukai K, Lee S, Matsudaira T, Taguchi T. A Role of Phosphatidylserine in the Function of Recycling Endosomes. Front Cell Dev Biol 2022; 9:783857. [PMID: 35004683 PMCID: PMC8740049 DOI: 10.3389/fcell.2021.783857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Cells internalize proteins and lipids in the plasma membrane (PM) and solutes in the extracellular space by endocytosis. The removal of PM by endocytosis is constantly balanced by the replenishment of proteins and lipids to PM through recycling pathway. Recycling endosomes (REs) are specific subsets of endosomes. Besides the established role of REs in recycling pathway, recent studies have revealed unanticipated roles of REs in membrane traffic and cell signalling. In this review, we highlight these emerging issues, with a particular focus on phosphatidylserine (PS), a phospholipid that is highly enriched in the cytosolic leaflet of RE membranes. We also discuss the pathogenesis of Hermansky Pudlak syndrome type 2 (HPS2) that arises from mutations in the AP3B1 gene, from the point of view of dysregulated RE functions.
Collapse
Affiliation(s)
- Junya Hasegawa
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yasunori Uchida
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Kojiro Mukai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Shoken Lee
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Tatsuyuki Matsudaira
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Tomohiko Taguchi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Kawasaki A, Sakai A, Nakanishi H, Hasegawa J, Taguchi T, Sasaki J, Arai H, Sasaki T, Igarashi M, Nakatsu F. PI4P/PS countertransport by ORP10 at ER-endosome membrane contact sites regulates endosome fission. J Cell Biol 2022; 221:212876. [PMID: 34817532 PMCID: PMC8624802 DOI: 10.1083/jcb.202103141] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/28/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Membrane contact sites (MCSs) serve as a zone for nonvesicular lipid transport by oxysterol-binding protein (OSBP)-related proteins (ORPs). ORPs mediate lipid countertransport, in which two distinct lipids are transported counterdirectionally. How such lipid countertransport controls specific biological functions, however, remains elusive. We report that lipid countertransport by ORP10 at ER–endosome MCSs regulates retrograde membrane trafficking. ORP10, together with ORP9 and VAP, formed ER–endosome MCSs in a phosphatidylinositol 4-phosphate (PI4P)-dependent manner. ORP10 exhibited a lipid exchange activity toward its ligands, PI4P and phosphatidylserine (PS), between liposomes in vitro, and between the ER and endosomes in situ. Cell biological analysis demonstrated that ORP10 supplies a pool of PS from the ER, in exchange for PI4P, to endosomes where the PS-binding protein EHD1 is recruited to facilitate endosome fission. Our study highlights a novel lipid exchange at ER–endosome MCSs as a nonenzymatic PI4P-to-PS conversion mechanism that organizes membrane remodeling during retrograde membrane trafficking.
Collapse
Affiliation(s)
- Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Akiko Sakai
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Hiroki Nakanishi
- Graduate School of Medicine and Research Center for Biosignal, Akita University, Akita, Japan
| | - Junya Hasegawa
- Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiko Taguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junko Sasaki
- Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Arai
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takehiko Sasaki
- Graduate School of Medicine and Research Center for Biosignal, Akita University, Akita, Japan.,Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Niigata University School of Medicine and Graduate School of Medical/Dental Sciences, Niigata, Japan
| |
Collapse
|
17
|
Gu H, Peng Y, Chen Y. An Emerging Therapeutic Approach by Targeting Myoferlin (MYOF) for Malignant Tumors. Curr Top Med Chem 2021; 20:1509-1515. [PMID: 32552653 DOI: 10.2174/1568026620666200618123436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/13/2020] [Accepted: 06/13/2020] [Indexed: 12/31/2022]
Abstract
Myoferlin (MYOF), as a member of the ferlin family, is a type II transmembrane protein with a single transmembrane domain at the carbon terminus. Studies have shown that MYOF is involved in pivotal physiological functions related to numerous cell membranes, such as extracellular secretion, endocytosis cycle, vesicle trafficking, membrane repair, membrane receptor recycling, and secreted protein efflux. Recently, the studies have also revealed that MYOF is overexpressed in a variety of cancers such as colorectal cancer, pancreatic cancer, breast cancer, melanoma, gastric cancer, and non-small-cell lung cancer. High expression of MYOF is associated with the high invasion of tumors and poor clinical prognosis. MYOF medicates the expression, secretion, and distribution of proteins, which were closely related to cancers, as well as the energy utilization of cancer cells, lipid metabolism and other physiological activities by regulating the physiological processes of membrane transport. In this short article, we briefly summarize the latest progress related to MYOF, indicating that small molecule inhibitors targeting the MYOF-C2D domain can selectively inhibit the proliferation and migration of cancer cells, and MYOF may be a promising target for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Haijun Gu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yangrui Peng
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
18
|
Lin YS, Huang KY, Yu HC, Lu MC, Fan CJ, Huang Tseng HY, Jhuang BY, Liu SQ, Lai NS, Lin TH, Huang HB. Identification of phostensin in association with Eps 15 homology domain-containing protein 1 (EHD1) and EHD4. Biochem Biophys Res Commun 2020; 531:236-241. [PMID: 32800345 DOI: 10.1016/j.bbrc.2020.07.030] [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: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 11/17/2022]
Abstract
Phostensin (PTS) encoded by KIAA1949 is a protein phosphatase 1 (PP1)-binding protein. In order to explore the cellular functions of PTS, we have searched PTS-binding proteins by using co-immunoprecipitation in combination with shotgun proteomics. Here, we report two novel PTS-binding proteins, Eps 15 homology domain-containing protein 1 (EHD1) and EHD4. PTS associated with EHD proteins was also observed in GST pull-down assays. Immunofluorescence microscopy demonstrated that the complex was co-localized at the endocytic vesicles. EHD proteins have been known to play a critical role in regulation of endocytic transport. Overexpression of PTS-β can attenuate the endocytic trafficking of transferrin.
Collapse
Affiliation(s)
- Yu-Shan Lin
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, 621, Taiwan
| | - Kuang-Yung Huang
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, 62247, Taiwan; School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Hui-Chun Yu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, 62247, Taiwan
| | - Ming-Chi Lu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, 62247, Taiwan; School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Cheng-Jhong Fan
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, 621, Taiwan
| | - Hsien-Yu Huang Tseng
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, 621, Taiwan
| | - Bi-Yao Jhuang
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, 621, Taiwan
| | - Su-Qin Liu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, 62247, Taiwan
| | - Ning-Sheng Lai
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, 62247, Taiwan; School of Medicine, Tzu Chi University, Hualien, 970, Taiwan.
| | - Ta-Hsien Lin
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, 11217, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan.
| | - Hsien-Bin Huang
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, 621, Taiwan.
| |
Collapse
|
19
|
Jones T, Naslavsky N, Caplan S. Eps15 Homology Domain Protein 4 (EHD4) is required for Eps15 Homology Domain Protein 1 (EHD1)-mediated endosomal recruitment and fission. PLoS One 2020; 15:e0239657. [PMID: 32966336 PMCID: PMC7511005 DOI: 10.1371/journal.pone.0239657] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/10/2020] [Indexed: 11/19/2022] Open
Abstract
Upon internalization, receptors are trafficked to sorting endosomes (SE) where they undergo sorting and are then packaged into budding vesicles that undergo fission and transport within the cell. Eps15 Homology Domain Protein 1 (EHD1), the best-characterized member of the Eps15 Homology Domain Protein (EHD) family, has been implicated in catalyzing the fission process that releases endosome-derived vesicles for recycling to the plasma membrane. Indeed, recent studies suggest that upon receptor-mediated internalization, EHD1 is recruited from the cytoplasm to endosomal membranes where it catalyzes vesicular fission. However, the mechanism by which this recruitment occurs remains unknown. Herein, we demonstrate that the EHD1 paralog, EHD4, is required for the recruitment of EHD1 to SE. We show that EHD4 preferentially dimerizes with EHD1, and knock-down of EHD4 expression by siRNA, shRNA or by CRISPR/Cas9 gene-editing leads to impaired EHD1 SE-recruitment and enlarged SE. Moreover, we demonstrate that at least 3 different asparagine-proline-phenylalanine (NPF) motif-containing EHD binding partners, Rabenosyn-5, Syndapin2 and MICAL-L1, are required for the recruitment of EHD1 to SE. Indeed, knock-down of any of these SE-localized EHD interaction partners leads to enlarged SE, presumably due to impaired endosomal fission. Overall, we identify a novel mechanistic role for EHD4 in recruitment of EHD1 to SE, thus positioning EHD4 as an essential component of the EHD1-fission machinery at SE.
Collapse
Affiliation(s)
- Tyler Jones
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Naava Naslavsky
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Steve Caplan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States of America
- * E-mail:
| |
Collapse
|
20
|
Bhattacharyya S, Pucadyil TJ. Cellular functions and intrinsic attributes of the ATP-binding Eps15 homology domain-containing proteins. Protein Sci 2020; 29:1321-1330. [PMID: 32223019 DOI: 10.1002/pro.3860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 01/14/2023]
Abstract
Several cellular processes rely on a cohort of dedicated proteins that manage tubulation, fission, and fusion of membranes. A notably large number of them belong to the dynamin superfamily of proteins. Among them is the evolutionarily conserved group of ATP-binding Eps15-homology domain-containing proteins (EHDs). In the two decades since their discovery, EHDs have been linked to a range of cellular processes that require remodeling or maintenance of specific membrane shapes such as during endocytic recycling, caveolar biogenesis, ciliogenesis, formation of T-tubules in skeletal muscles, and membrane resealing after rupture. Recent work has shed light on their structure and the unique attributes they possess in linking ATP hydrolysis to membrane remodeling. This review summarizes some of these recent developments and reconciles intrinsic protein functions to their cellular roles.
Collapse
Affiliation(s)
- Soumya Bhattacharyya
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Thomas J Pucadyil
- Department of Biology, Indian Institute of Science Education and Research, Pune, India
| |
Collapse
|
21
|
Dhawan K, Naslavsky N, Caplan S. Sorting nexin 17 (SNX17) links endosomal sorting to Eps15 homology domain protein 1 (EHD1)-mediated fission machinery. J Biol Chem 2020; 295:3837-3850. [PMID: 32041776 DOI: 10.1074/jbc.ra119.011368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Following endocytosis, receptors that are internalized to sorting endosomes are sorted to different pathways, in part by sorting nexin (SNX) proteins. Notably, SNX17 interacts with a multitude of receptors in a sequence-specific manner to regulate their recycling. However, the mechanisms by which SNX17-labeled vesicles that contain sorted receptors bud and undergo vesicular fission from the sorting endosomes remain elusive. Recent studies suggest that a dynamin-homolog, Eps15 homology domain protein 1, catalyzes fission and releases endosome-derived vesicles for recycling to the plasma membrane. However, the mechanism by which EHD1 is coupled to various receptors and regulates their recycling remains unknown. Here we sought to characterize the mechanism by which EHD1 couples with SNX17 to regulate recycling of SNX17-interacting receptors. We hypothesized that SNX17 couples receptors to the EHD1 fission machinery in mammalian cells. Coimmunoprecipitation experiments and in vitro assays provided evidence that EHD1 and SNX17 directly interact. We also found that inducing internalization of a SNX17 cargo receptor, low-density lipoprotein receptor-related protein 1 (LRP1), led to recruitment of cytoplasmic EHD1 to endosomal membranes. Moreover, surface rendering and quantification of overlap volumes indicated that SNX17 and EHD1 partially colocalize on endosomes and that this overlap further increases upon LRP1 internalization. Additionally, SNX17-containing endosomes were larger in EHD1-depleted cells than in WT cells, suggesting that EHD1 depletion impairs SNX17-mediated endosomal fission. Our findings help clarify our current understanding of endocytic trafficking, providing significant additional insight into the process of endosomal fission and connecting the sorting and fission machineries.
Collapse
Affiliation(s)
- Kanika Dhawan
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center, Omaha, Nebraska 68198 .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|
22
|
Zhu W, Zhou B, Zhao C, Ba Z, Xu H, Yan X, Liu W, Zhu B, Wang L, Ren C. Myoferlin, a multifunctional protein in normal cells, has novel and key roles in various cancers. J Cell Mol Med 2019; 23:7180-7189. [PMID: 31475450 PMCID: PMC6815776 DOI: 10.1111/jcmm.14648] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/30/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
Myoferlin, a protein of the ferlin family, has seven C2 domains and exhibits activity in some cells, including myoblasts and endothelial cells. Recently, myoferlin was identified as a promising target and biomarker in non-small-cell lung cancer, breast cancer, pancreatic adenocarcinoma, hepatocellular carcinoma, colon cancer, melanoma, oropharyngeal squamous cell carcinoma, head and neck squamous cell carcinoma, clear cell renal cell carcinoma and endometrioid carcinoma. This evidence indicated that myoferlin was involved in the proliferation, invasion and migration of tumour cells, the mechanism of which mainly included promoting angiogenesis, vasculogenic mimicry, energy metabolism reprogramming, epithelial-mesenchymal transition and modulating exosomes. The roles of myoferlin in both normal cells and cancer cells are of great significance to provide novel and efficient methods of tumour treatment. In this review, we summarize recent studies and findings of myoferlin and suggest that myoferlin is a novel potential candidate for clinical diagnosis and targeted cancer therapy.
Collapse
Affiliation(s)
- Wei Zhu
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Bolun Zhou
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Chenxuan Zhao
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zhengqing Ba
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hongjuan Xu
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xuejun Yan
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Weidong Liu
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Bin Zhu
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Lei Wang
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Caiping Ren
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Piechulek A, Berwanger LC, von Mikecz A. Silica nanoparticles disrupt OPT-2/PEP-2-dependent trafficking of nutrient peptides in the intestinal epithelium. Nanotoxicology 2019; 13:1133-1148. [DOI: 10.1080/17435390.2019.1643048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Annette Piechulek
- IUF – Leibniz Research Institute for Environmental Medicine, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Lutz C. Berwanger
- IUF – Leibniz Research Institute for Environmental Medicine, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Anna von Mikecz
- IUF – Leibniz Research Institute for Environmental Medicine, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
24
|
Deo R, Kushwah MS, Kamerkar SC, Kadam NY, Dar S, Babu K, Srivastava A, Pucadyil TJ. ATP-dependent membrane remodeling links EHD1 functions to endocytic recycling. Nat Commun 2018; 9:5187. [PMID: 30518883 PMCID: PMC6281616 DOI: 10.1038/s41467-018-07586-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 11/02/2018] [Indexed: 01/30/2023] Open
Abstract
Endocytic and recycling pathways generate cargo-laden transport carriers by membrane fission. Classical dynamins, which generate transport carriers during endocytosis, constrict and cause fission of membrane tubes in response to GTP hydrolysis. Relatively, less is known about the ATP-binding Eps15-homology domain-containing protein1 (EHD1), a dynamin family member that functions at the endocytic-recycling compartment. Here, we show using cross complementation assays in C. elegans that EHD1's membrane binding and ATP hydrolysis activities are necessary for endocytic recycling. Further, we show that ATP-bound EHD1 forms membrane-active scaffolds that bulge tubular model membranes. ATP hydrolysis promotes scaffold self-assembly, causing the bulge to extend and thin down intermediate regions on the tube. On tubes below 25 nm in radius, such thinning leads to scission. Molecular dynamics simulations corroborate this scission pathway. Deletion of N-terminal residues causes defects in stable scaffolding, scission and endocytic recycling. Thus, ATP hydrolysis-dependent membrane remodeling links EHD1 functions to endocytic recycling.
Collapse
Affiliation(s)
- Raunaq Deo
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Manish S Kushwah
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Sukrut C Kamerkar
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Nagesh Y Kadam
- Indian Institute of Science Education and Research, Sector 81, S.A.S Nagar, Mohali, 140306, Punjab, India
| | - Srishti Dar
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Kavita Babu
- Indian Institute of Science Education and Research, Sector 81, S.A.S Nagar, Mohali, 140306, Punjab, India
| | - Anand Srivastava
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Thomas J Pucadyil
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India.
| |
Collapse
|
25
|
Lučin P, Kareluša L, Blagojević Zagorac G, Mahmutefendić Lučin H, Pavišić V, Jug Vučko N, Lukanović Jurić S, Marcelić M, Lisnić B, Jonjić S. Cytomegaloviruses Exploit Recycling Rab Proteins in the Sequential Establishment of the Assembly Compartment. Front Cell Dev Biol 2018; 6:165. [PMID: 30564576 PMCID: PMC6288171 DOI: 10.3389/fcell.2018.00165] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/19/2018] [Indexed: 12/19/2022] Open
Abstract
Cytomegaloviruses (CMV) reorganize membranous system of the cell in order to develop a virion assembly compartment (VAC). The development starts in the early (E) phase of infection with the reorganization of the endosomal system and the Golgi and proceeds to the late phase until newly formed virions are assembled and released. The events in the E phase involve reorganization of the endosomal recycling compartment (ERC) in a series of cellular alterations that are mostly unknown. In this minireview, we discuss the effect of murine CMV infection on Rab proteins, master regulators of membrane trafficking pathways, which in the cascades with their GEFs and GAPs organize the flow of membranes through the ERC. Immunofluorescence analyzes of murine CMV infected cells suggest perturbations of Rab cascades that operate at the ERC. Analysis of cellular transcriptome in the course of both murine and human CMV infection demonstrates the alteration in expression of cellular genes whose products are known to build Rab cascades. These alterations, however, cannot explain perturbations of the ERC. Cellular proteome data available for human CMV infected cells suggests the potential role of RabGAP downregulation at the end of the E phase. However, the very early onset of the ERC alterations in the course of MCMV infection indicates that CMVs exploit Rab cascades to reorganize the ERC, which represents the earliest step in the sequential establishment of the cVAC.
Collapse
Affiliation(s)
- Pero Lučin
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,University North - University Center Varaždin, Varaždin, Croatia
| | - Ljerka Kareluša
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Hana Mahmutefendić Lučin
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.,University North - University Center Varaždin, Varaždin, Croatia
| | - Valentino Pavišić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Natalia Jug Vučko
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Silvija Lukanović Jurić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marina Marcelić
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
26
|
Kamerkar SC, Roy K, Bhattacharyya S, Pucadyil TJ. A Screen for Membrane Fission Catalysts Identifies the ATPase EHD1. Biochemistry 2018; 58:65-71. [PMID: 30403133 PMCID: PMC6327249 DOI: 10.1021/acs.biochem.8b00925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Membrane fission manifests during cell division, synaptic transmission, vesicular transport, and organelle biogenesis, yet identifying proteins that catalyze fission remains a challenge. Using a facile and robust assay system of supported membrane tubes in a microscopic screen that directly monitors membrane tube scission, we detect robust GTP- and ATP-dependent as well as nucleotide-independent fission activity in the brain cytosol. Using previously established interacting partner proteins as bait for pulldowns, we attribute the GTP-dependent fission activity to dynamin. Biochemical fractionation followed by mass spectrometric analyses identifies the Eps15-homology domain-containing protein1 (EHD1) as a novel ATP-dependent membrane fission catalyst. Together, our approach establishes an experimental workflow for the discovery of novel membrane fission catalysts.
Collapse
Affiliation(s)
- Sukrut C Kamerkar
- Indian Institute of Science Education and Research , Dr. Homi Bhabha Road , Pashan, Pune 411008 , Maharashtra , India
| | - Krishnendu Roy
- Indian Institute of Science Education and Research , Dr. Homi Bhabha Road , Pashan, Pune 411008 , Maharashtra , India
| | - Soumya Bhattacharyya
- Indian Institute of Science Education and Research , Dr. Homi Bhabha Road , Pashan, Pune 411008 , Maharashtra , India
| | - Thomas J Pucadyil
- Indian Institute of Science Education and Research , Dr. Homi Bhabha Road , Pashan, Pune 411008 , Maharashtra , India
| |
Collapse
|
27
|
Wu C, Bao G, Xu G, Sun Y, Wang L, Chen J, Zhang J, Chen C, Zhu Q, Cui Z. Triad1 regulates the expression and distribution of EHD1 contributing to the neurite outgrowth of neurons after spinal cord injury. J Cell Biochem 2018; 120:5355-5366. [PMID: 30320922 DOI: 10.1002/jcb.27814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/12/2018] [Indexed: 12/18/2022]
Abstract
Traumatic spinal cord injury is a common and severe complication after an accident. As we all know that neurite outgrowth of neurons is difficult after a spinal cord injury. Endosome system is associated with cargoes transportation and contributes in promoting the neuronal capability for neurite outgrowth. EH domain-containing protein 1 (EHD1) transports proteins through the endosome system, especially in the recycling endosomes and regulating the neurite outgrowth. In mammalian cells, the involvement of the ubiquitin-proteasome system in endosomal sorting has been well established. Two RING fingers and a DRIL (double RING finger-linked) 1 (Triad1) plays an important role in membrane trafficking and its mutant results in the wrong accumulation of receptors in endosomes and plasma membrane. In this current study, we reasonably integrated the results of the above research and investigated the regulating function of Triad1 to EHD1 following the spinal cord injury. We characterized the upregulated expression and distribution of Triad1 and EHD1 in the neurons after SCI and declared the interaction between Triad1 with EHD1 both in vitro and in vivo. Triad1 regulated the interaction between itself and the full-length or EH domain of EHD1, which influenced the neurite outgrowth of PC12 cells. Our data delineate a novel interaction between Triad1 and EHD1 that may contribute to the regulation of neurite outgrowth for neurons after the spinal cord injury.
Collapse
Affiliation(s)
- Chunshuai Wu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Guofeng Bao
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Guanhua Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Yuyu Sun
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Lingling Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Jiajia Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Jinlong Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Chu Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Qiancheng Zhu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
28
|
Liu Y, Liang Y, Li M, Liu D, Tang J, Yang W, Tong D, Jin X. Eps15 homology domain 1 promotes the evolution of papillary thyroid cancer by regulating endocytotic recycling of epidermal growth factor receptor. Oncol Lett 2018; 16:4263-4270. [PMID: 30214560 PMCID: PMC6126170 DOI: 10.3892/ol.2018.9200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of thyroid malignancy, and it is often observed to overexpress epidermal growth factor receptor (EGFR). Previous research has indicated that EH domain-containing 1 (EHD1) is associated with EGFR-mediated endocytotic recycling in multiple tumor types. The objective of the present study was to determine the protein expression levels and clinical significance of EHD1, EGFR, caveolin-1 (CAV-1) and RAB11 family interacting protein 3 (RAB11FIP3) in PTC. PTC specimens were analyzed for EHD1, EGFR, CAV-1 and RAB11FIP3 expression via immunohistochemistry and western blotting. The associations between protein expression and clinicopathological features were assessed. EHD1, EGFR, CAV-1 and RAB11FIP3 expression levels were increased in human PTC. Additionally, the expression level of EHD1 protein was significantly associated with tumor size, lymph node metastasis and EGFR expression (P<0.05). CAV-1 was associated with tumor size and EGFR expression (P<0.05). EGFR was only associated with lymph node metastasis (P=0.027) and RAB11FIP3 was not associated with any clinicopathological characteristics. The correlations between EHD1 and EGFR (r=0.564, P<0.05), CAV-1 (r=0.865, P<0.01) and RAB11FIP3 (r=0.504, P<0.05) were statistically significant. Overall, EHD1, CAV-1 and RAB11FIP3, which are key proteins in endocytotic recycling, promote PTC tumorigenesis through the regulation of the transport of EGFR.
Collapse
Affiliation(s)
- Yu Liu
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yanan Liang
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China.,College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Ming Li
- Institute of Iodine Deficiency Disorders, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Duanyang Liu
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jing Tang
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Weiwei Yang
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dandan Tong
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoming Jin
- Department of Pathology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
29
|
Abstract
Antigen cross-presentation is an adaptation of the cellular process of loading MHC-I molecules with endogenous peptides during their biosynthesis within the endoplasmic reticulum. Cross-presented peptides derive from internalized proteins, microbial pathogens, and transformed or dying cells. The physical separation of internalized cargo from the endoplasmic reticulum, where the machinery for assembling peptide-MHC-I complexes resides, poses a challenge. To solve this problem, deliberate rewiring of organelle communication within cells is necessary to prepare for cross-presentation, and different endocytic receptors and vesicular traffic patterns customize the emergent cross-presentation compartment to the nature of the peptide source. Three distinct pathways of vesicular traffic converge to form the ideal cross-presentation compartment, each regulated differently to supply a unique component that enables cross-presentation of a diverse repertoire of peptides. Delivery of centerpiece MHC-I molecules is the critical step regulated by microbe-sensitive Toll-like receptors. Defining the subcellular sources of MHC-I and identifying sites of peptide loading during cross-presentation remain key challenges.
Collapse
Affiliation(s)
- J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; .,Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
30
|
Spatio-temporal regulation of EGFR signaling by the Eps15 homology domain-containing protein 3 (EHD3). Oncotarget 2018; 7:79203-79216. [PMID: 27811356 PMCID: PMC5346708 DOI: 10.18632/oncotarget.13008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 08/21/2016] [Indexed: 11/25/2022] Open
Abstract
The epidermal growth factor (EGF) receptor EGFR is a major receptor tyrosine kinase whose role in gliomagenesis is well established. We have recently identified EHD3 [Eps15 homology (EH) domain-containing protein 3], an endocytic trafficking regulatory protein, as a putative brain tumor suppressor. Here, we investigate the underlying mechanisms, by establishing a novel mechanistic and functional connection between EHD3 and the EGFR signaling pathway. We show that, in response to stimulation with the EGF ligand, EHD3 accelerates the rate of EGFR degradation by dramatically increasing its ubiquitination. As part of this process, EHD3 also regulates EGFR endosomal trafficking by diverting it away from the recycling route into the degradative pathway. Moreover, we found that upon EGF activation, rather than affecting the total MAPK and AKT downstream signaling, EHD3 decreases endosome-based signaling of these two pathways, thus suggesting the contribution of EHD3 in the spatial regulation of EGFR signaling. This function explains the higher sensitivity of EHD3-expressing cells to the growth-inhibitory effects of EGF. In summary, this is the first report supporting a mechanism of EHD3-mediated tumor suppression that involves the attenuation of endosomal signaling of the EGFR oncogene.
Collapse
|
31
|
Rahman SS, Moffitt AEJ, Trease AJ, Foster KW, Storck MD, Band H, Boesen EI. EHD4 is a novel regulator of urinary water homeostasis. FASEB J 2017; 31:5217-5233. [PMID: 28778975 DOI: 10.1096/fj.201601182rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 07/25/2017] [Indexed: 01/08/2023]
Abstract
The Eps15-homology domain-containing (EHD) protein family comprises 4 members that regulate endocytic recycling. Although the kidney expresses all 4 EHD proteins, their physiologic roles are largely unknown. This study focused on EHD4, which we found to be expressed differentially across nephron segments with the highest expression in the inner medullary collecting duct. Under baseline conditions, Ehd4-/- [EHD4-knockout (KO)] mice on a C57Bl/6 background excreted a higher volume of more dilute urine than control C57Bl/6 wild-type (WT) mice while maintaining a similar plasma osmolality. Urine excretion after an acute intraperitoneal water load was significantly increased in EHD4-KO mice compared to WT mice, and although EHD4-KO mice concentrated their urine during 24-h water restriction, urinary osmolality remained significantly lower than in WT mice, suggesting that EHD4 plays a role in renal water handling. Total aquaporin 2 (AQP2) and phospho-S256-AQP2 (pAQP2) protein expression in the inner medulla was similar in the two groups in baseline conditions. However, localization of both AQP2 and pAQP2 in the renal inner medullary principal cells appeared more dispersed, and the intensity of apical membrane staining for AQP2 was reduced significantly (by ∼20%) in EHD4-KO mice compared to WT mice in baseline conditions, suggesting an important role of EHD4 in trafficking of AQP2. Together, these data indicate that EHD4 play important roles in the regulation of water homeostasis.-Rahman, S. S., Moffitt, A. E. J., Trease, A. J., Foster, K. W., Storck, M. D., Band, H., Boesen, E. I. EHD4 is a novel regulator of urinary water homeostasis.
Collapse
Affiliation(s)
- Shamma S Rahman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Alexandra E J Moffitt
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew D Storck
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Hamid Band
- The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska, USA; .,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA; and.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| |
Collapse
|
32
|
Blander JM. The comings and goings of MHC class I molecules herald a new dawn in cross-presentation. Immunol Rev 2017; 272:65-79. [PMID: 27319343 DOI: 10.1111/imr.12428] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MHC class I (MHC-I) molecules are the centerpieces of cross-presentation. They are loaded with peptides derived from exogenous sources and displayed on the plasma membrane to communicate with CD8 T cells, relaying a message of tolerance or attack. The study of cross-presentation has been focused on the relative contributions of the vacuolar versus cytosolic pathways of antigen processing and the location where MHC-I molecules are loaded. While vacuolar processing generates peptides loaded onto vacuolar MHC-I molecules, how and where exogenous peptides generated by the proteasome and transported by TAP meet MHC-I molecules for loading has been a matter of debate. The source and trafficking of MHC-I molecules in dendritic cells have largely been ignored under the expectation that these molecules came from the Endoplasmic reticulum (ER) or the plasma membrane. New studies reveal a concentrated pool of MHC-I molecules in the endocytic recycling compartment (ERC). These pools are rapidly mobilized to phagosomes carrying microbial antigens, and in a signal-dependent manner under the control of Toll-like receptors. The phagosome becomes a dynamic hub receiving traffic from multiple sources, the ER-Golgi intermediate compartment for delivering the peptide-loading machinery and the ERC for deploying MHC-I molecules that alert CD8 T cells of infection.
Collapse
Affiliation(s)
- J Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
33
|
Abstract
Cross-presentation of internalized antigens by dendritic cells requires efficient delivery of Major Histocompatibility Complex (MHC) class I molecules to peptide-loading compartments. Strong evidence suggests that such loading can occur outside of the endoplasmic reticulum; however, the trafficking pathways and sources of class I molecules involved are poorly understood. Examination of non-professional, non-phagocytic cells has revealed a clathrin-independent, Arf6-dependent recycling pathway likely traveled by internalized optimally loaded (closed) class I molecules. Some closed and all open MHC class I molecules travel to late endosomes to be degraded but might also partly be re-loaded with peptides and recycled. Studies of viral interference revealed pathways in which class I molecules are directed to degradation in lysosomes upon ubiquitination at the surface, or upon AP-1 and HIV-nef-dependent misrouting from the Golgi network to lysosomes. While many observations made in non-professional cells remain to be re-examined in dendritic cells, available evidence suggests that both recycling and neo-synthesized class I molecules can be loaded with cross-presented peptides. Recycling molecules can be recruited to phagosomes triggered by innate signals such as TLR4 ligands, and may therefore specialize in loading with phagocytosed antigens. In contrast, AP-1-dependent accumulation at, or trafficking through, a Golgi compartment of newly synthesized molecules appears to be important for cross-presentation of soluble proteins and possibly of long peptides that are processed in the so-called vacuolar pathway. However, significant cell biological work will be required to confirm this or any other model and to integrate knowledge on MHC class I biochemistry and trafficking in models of CD8(+) T-cell priming by dendritic cells.
Collapse
Affiliation(s)
- Peter van Endert
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Centre National de la Recherche Scientifique, Unité 8253, Paris, France
| |
Collapse
|
34
|
Tong D, Liang YN, Stepanova AA, Liu Y, Li X, Wang L, Zhang F, Vasilyeva NV. Increased Eps15 homology domain 1 and RAB11FIP3 expression regulate breast cancer progression via promoting epithelial growth factor receptor recycling. Tumour Biol 2017; 39:1010428317691010. [DOI: 10.1177/1010428317691010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recent research indicates that the C-terminal Eps15 homology domain 1 is associated with epithelial growth factor receptor–mediated endocytosis recycling in non-small-cell lung cancer. The aim of this study was to determine the clinical significance of Eps15 homology domain 1 gene expression in relation to phosphorylation of epithelial growth factor receptor expression in patients with breast cancer. Primary breast cancer samples from 306 patients were analyzed for Eps15 homology domain 1, RAB11FIP3, and phosphorylation of epithelial growth factor receptor expression via immunohistochemistry. The clinical significance was assessed via a multivariate Cox regression analysis, Kaplan–Meier curves, and the log-rank test. Eps15 homology domain 1 and phosphorylation of epithelial growth factor receptor were upregulated in 60.46% (185/306) and 53.92% (165/306) of tumor tissues, respectively, as assessed by immunohistochemistry. The statistical correlation analysis indicated that Eps15 homology domain 1 overexpression was positively correlated with the increases in phosphorylation of epithelial growth factor receptor ( r = 0.242, p < 0.001) and RAB11FIP3 ( r = 0.165, p = 0.005) expression. The multivariate Cox proportional hazard model analysis demonstrated that the expression of Eps15 homology domain 1 alone is a significant prognostic marker of breast cancer for the overall survival in the total, chemotherapy, and human epidermal growth factor receptor 2 (−) groups. However, the use of combined expression of Eps15 homology domain 1 and phosphorylation of epithelial growth factor receptor markers is more effective for the disease-free survival in the overall population, chemotherapy, and human epidermal growth factor receptor 2 (−) groups. Moreover, the combined markers are also significant prognostic markers of breast cancer in the human epidermal growth factor receptor 2 (+), estrogen receptor (+), and estrogen receptor (−) groups. Eps15 homology domain 1 has a tumor suppressor function, and the combined marker of Eps15 homology domain 1/phosphorylation of epithelial growth factor receptor expression was identified as a better prognostic marker in breast cancer diagnosis. Furthermore, RAB11FIP3 combines with Eps15 homology domain 1 to promote the endocytosis recycling of phosphorylation of epithelial growth factor receptor.
Collapse
Affiliation(s)
- Dandan Tong
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Ya-Nan Liang
- Department of Pathology, Harbin Medical University, Harbin, China
- College of Pharmacy, Harbin Medical University, Harbin, China
| | - AA Stepanova
- Kashkin Research Institute of Medical Mycology, I.I. Mechnikov North-Western State Medical University, Saint Petersburg, Russia
| | - Yu Liu
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Letian Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fengmin Zhang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - NV Vasilyeva
- Kashkin Research Institute of Medical Mycology, I.I. Mechnikov North-Western State Medical University, Saint Petersburg, Russia
| |
Collapse
|
35
|
Yu H, Wang MJ, Xuan NX, Shang ZC, Wu J. Molecular dynamics simulation of the interactions between EHD1 EH domain and multiple peptides. J Zhejiang Univ Sci B 2016; 16:883-96. [PMID: 26465136 DOI: 10.1631/jzus.b1500106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To provide essential information for peptide inhibitor design, the interactions of Eps15 homology domain of Eps15 homology domain-containing protein 1 (EHD1 EH domain) with three peptides containing NPF (asparagine-proline-phenylalanine), DPF (aspartic acid-proline-phenylalanine), and GPF (glycine-proline-phenylalanine) motifs were deciphered at the atomic level. The binding affinities and the underlying structure basis were investigated. METHODS Molecular dynamics (MD) simulations were performed on EHD1 EH domain/peptide complexes for 60 ns using the GROMACS package. The binding free energies were calculated and decomposed by molecular mechanics/generalized Born surface area (MM/GBSA) method using the AMBER package. The alanine scanning was performed to evaluate the binding hot spot residues using FoldX software. RESULTS The different binding affinities for the three peptides were affected dominantly by van der Waals interactions. Intermolecular hydrogen bonds provide the structural basis of contributions of van der Waals interactions of the flanking residues to the binding. CONCLUSIONS van der Waals interactions should be the main consideration when we design peptide inhibitors of EHD1 EH domain with high affinities. The ability to form intermolecular hydrogen bonds with protein residues can be used as the factor for choosing the flanking residues.
Collapse
Affiliation(s)
- Hua Yu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Mao-jun Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Nan-xia Xuan
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Zhi-cai Shang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Jun Wu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
36
|
Wu C, Cui Z, Liu Y, Zhang J, Ding W, Wang S, Bao G, Xu G, Sun Y, Chen J. The importance of EHD1 in neurite outgrowth contributing to the functional recovery after spinal cord injury. Int J Dev Neurosci 2016; 52:24-32. [PMID: 27211346 DOI: 10.1016/j.ijdevneu.2016.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/11/2016] [Accepted: 05/18/2016] [Indexed: 11/15/2022] Open
Affiliation(s)
- Chunshuai Wu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Yonghua Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jinlong Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China
| | - Wensen Ding
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Song Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Guofeng Bao
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China
| | - Guanhua Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuyu Sun
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China
| | - Jiajia Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Haier Lane North Road No. 6, Nantong, 226001, Jiangsu, People's Republic of China
| |
Collapse
|
37
|
Cypher LR, Bielecki TA, Adepegba O, Huang L, An W, Iseka F, Luan H, Tom E, Storck MD, Hoppe AD, Band V, Band H. CSF-1 receptor signalling is governed by pre-requisite EHD1 mediated receptor display on the macrophage cell surface. Cell Signal 2016; 28:1325-1335. [PMID: 27224507 DOI: 10.1016/j.cellsig.2016.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/23/2022]
Abstract
Colony stimulating factor-1 receptor (CSF-1R), a receptor tyrosine kinase (RTK), is the master regulator of macrophage biology. CSF-1 can bind CSF-1R resulting in receptor activation and signalling essential for macrophage functions such as proliferation, differentiation, survival, polarization, phagocytosis, cytokine secretion, and motility. CSF-1R activation can only occur after the receptor is presented on the macrophage cell surface. This process is reliant upon the underlying macrophage receptor trafficking machinery. However, the mechanistic details governing this process are incompletely understood. C-terminal Eps15 Homology Domain-containing (EHD) proteins have recently emerged as key regulators of receptor trafficking but have not yet been studied in the context of macrophage CSF-1R signalling. In this manuscript, we utilize primary bone-marrow derived macrophages (BMDMs) to reveal a novel function of EHD1 as a regulator of CSF-1R abundance on the cell surface. We report that EHD1-knockout (EHD1-KO) macrophages cell surface and total CSF-1R levels are significantly decreased. The decline in CSF-1R levels corresponds with reduced downstream macrophage functions such as cell proliferation, migration, and spreading. In EHD1-KO macrophages, transport of newly synthesized CSF-1R to the macrophage cell surface was reduced and was associated with the shunting of the receptor to the lysosome, which resulted in receptor degradation. These findings reveal a novel and functionally important role for EHD1 in governing CSF-1R signalling via regulation of anterograde transport of CSF-1R to the macrophage cell surface.
Collapse
Affiliation(s)
- Luke R Cypher
- Eppley Cancer Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Timothy Alan Bielecki
- Eppley Cancer Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | | | - Lu Huang
- Department of Chemistry and Biochemistry, BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Wei An
- Department of Genetics, Cell Biology, & Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fany Iseka
- Department of Genetics, Cell Biology, & Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | | | - Eric Tom
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Matthew D Storck
- Eppley Cancer Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam D Hoppe
- Department of Chemistry and Biochemistry, BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Vimla Band
- Eppley Cancer Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States; Department of Genetics, Cell Biology, & Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hamid Band
- Eppley Cancer Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States; Department of Genetics, Cell Biology, & Anatomy, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
38
|
Bahl K, Xie S, Spagnol G, Sorgen P, Naslavsky N, Caplan S. EHD3 Protein Is Required for Tubular Recycling Endosome Stabilization, and an Asparagine-Glutamic Acid Residue Pair within Its Eps15 Homology (EH) Domain Dictates Its Selective Binding to NPF Peptides. J Biol Chem 2016; 291:13465-78. [PMID: 27189942 DOI: 10.1074/jbc.m116.716407] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Indexed: 11/06/2022] Open
Abstract
An elaborate network of dynamic lipid membranes, termed tubular recycling endosomes (TRE), coordinates the process of endocytic recycling in mammalian cells. The C-terminal Eps15 homology domain (EHD)-containing proteins have been implicated in the bending and fission of TRE, thus regulating endocytic recycling. EHD proteins have an EH domain that interacts with proteins containing an NPF motif. We found that NPF-containing EHD1 interaction partners such as molecules interacting with CasL-like1 (MICAL-L1) and Syndapin2 are essential for TRE biogenesis. Also crucial for TRE biogenesis is the generation of phosphatidic acid, an essential lipid component of TRE that serves as a docking point for MICAL-L1 and Syndapin2. EHD1 and EHD3 have 86% amino acid identity; they homo- and heterodimerize and partially co-localize to TRE. Despite their remarkable identity, they have distinct mechanistic functions. EHD1 induces membrane vesiculation, whereas EHD3 supports TRE biogenesis and/or stabilization by an unknown mechanism. While using phospholipase D inhibitors (which block the conversion of glycerophospholipids to phosphatidic acid) to deplete cellular TRE, we observed that, upon inhibitor washout, there was a rapid and dramatic regeneration of MICAL-L1-marked TRE. Using this "synchronized" TRE biogenesis system, we determined that EHD3 is involved in the stabilization of TRE rather than in their biogenesis. Moreover, we identify the residues Ala-519/Asp-520 of EHD1 and Asn-519/Glu-520 of EHD3 as defining the selectivity of these two paralogs for NPF-containing binding partners, and we present a model to explain the atomic mechanism and provide new insight for their differential roles in vesiculation and tubulation, respectively.
Collapse
Affiliation(s)
- Kriti Bahl
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| | - Shuwei Xie
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| | - Gaelle Spagnol
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| | - Paul Sorgen
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| | - Naava Naslavsky
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| | - Steve Caplan
- From the Department of Biochemistry and Molecular Biology and Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870
| |
Collapse
|
39
|
Bhattacharyya S, Rainey MA, Arya P, Mohapatra BC, Mushtaq I, Dutta S, George M, Storck MD, McComb RD, Muirhead D, Todd GL, Gould K, Datta K, Gelineau-van Waes J, Band V, Band H. Endocytic recycling protein EHD1 regulates primary cilia morphogenesis and SHH signaling during neural tube development. Sci Rep 2016; 6:20727. [PMID: 26884322 PMCID: PMC4756679 DOI: 10.1038/srep20727] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/11/2016] [Indexed: 12/20/2022] Open
Abstract
Members of the four-member C-terminal EPS15-Homology Domain-containing (EHD) protein family play crucial roles in endocytic recycling of cell surface receptors from endosomes to the plasma membrane. In this study, we show that Ehd1 gene knockout in mice on a predominantly B6 background is embryonic lethal. Ehd1-null embryos die at mid-gestation with a failure to complete key developmental processes including neural tube closure, axial turning and patterning of the neural tube. We found that Ehd1-null embryos display short and stubby cilia on the developing neuroepithelium at embryonic day 9.5 (E9.5). Loss of EHD1 also deregulates the ciliary SHH signaling with Ehd1-null embryos displaying features indicative of increased SHH signaling, including a significant downregulation in the formation of the GLI3 repressor and increase in the ventral neuronal markers specified by SHH. Using Ehd1-null MEFS we found that EHD1 protein co-localizes with the SHH receptor Smoothened in the primary cilia upon ligand stimulation. Under the same conditions, EHD1 was shown to co-traffic with Smoothened into the developing primary cilia and we identify EHD1 as a direct binding partner of Smoothened. Overall, our studies identify the endocytic recycling regulator EHD1 as a novel regulator of the primary cilium-associated trafficking of Smoothened and Hedgehog signaling.
Collapse
Affiliation(s)
- Sohinee Bhattacharyya
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark A Rainey
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Priyanka Arya
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Samikshan Dutta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA
| | - Rodney D McComb
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Muirhead
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gordon L Todd
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Karen Gould
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaustubh Datta
- The Department of Biochemistry &Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Vimla Band
- The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hamid Band
- The Department of Pathology &Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,The Department of Genetics, Cell Biology &Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases,University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
40
|
Gueho A, Bosmani C, Gopaldass N, Molle V, Soldati T, Letourneur F. Dictyostelium EHD associates with Dynamin and participates in phagosome maturation. J Cell Sci 2016; 129:2354-67. [DOI: 10.1242/jcs.182857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 05/04/2016] [Indexed: 12/20/2022] Open
Abstract
C-terminal EHDs (Eps15 homology-domain-containing proteins) are newly identified key regulators of endosomal membrane trafficking. Here we show that D. discoideum contains a single EHD protein that localizes to endosomal compartments and newly formed phagosomes. We provide the first evidence that EHD regulates phagosome maturation. Deletion of EHD results in defects in intraphagosomal proteolysis and acidification. These defects are linked to early delivery of lysosomal enzymes and fast retrieval of the vacuolar H+-ATPase in maturing phagosomes. We also demonstrate that EHD physically interacts with DymA. Our results indicate that EHD and DymA can associate independently to endomembranes, and yet they share identical kinetics of phagosome recruitment and release during phagosome maturation. Functional analysis of ehd−, dymA−, and double dymA−/ehd− knock-out strains indicate that DymA and EHD play non-redundant and independent functions in phagosome maturation. Finally, we show that the absence of EHD leads to increase tubulation of endosomes, indicating that EHD participates in the scission of endosomal tubules as reported for DymA.
Collapse
Affiliation(s)
- Aurélie Gueho
- Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Cristina Bosmani
- Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Navin Gopaldass
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Virginie Molle
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS, UMR 5235, Place Eugène Bataillon, 34095 Montpellier Cedex 05, France
| | - Thierry Soldati
- Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - François Letourneur
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques, Université de Montpellier, CNRS, UMR 5235, Place Eugène Bataillon, 34095 Montpellier Cedex 05, France
| |
Collapse
|
41
|
Abstract
Since an intact membrane is required for normal cellular homeostasis, membrane repair is essential for cell survival. Human genetic studies, combined with the development of novel animal models and refinement of techniques to study cellular injury, have now uncovered series of repair proteins highly relevant for human health. Many of the deficient repair pathways manifest in skeletal muscle, where defective repair processes result in myopathies or other forms of muscle disease. Dysferlin is a membrane-associated protein implicated in sarcolemmal repair and also linked to other membrane functions including the maintenance of transverse tubules in muscle. MG53, annexins, and Eps15 homology domain-containing proteins interact with dysferlin to form a membrane repair complex and similarly have roles in membrane trafficking in muscle. These molecular features of membrane repair are not unique to skeletal muscle, but rather skeletal muscle, due to its high demands, is more dependent on an efficient repair process. Phosphatidylserine and phosphatidylinositol 4,5-bisphosphate, as well as Ca(2+), are central regulators of membrane organization during repair. Given the importance of muscle health in disease and in aging, these pathways are targets to enhance muscle function and recovery from injury.
Collapse
|
42
|
Arya P, Rainey MA, Bhattacharyya S, Mohapatra BC, George M, Kuracha MR, Storck MD, Band V, Govindarajan V, Band H. The endocytic recycling regulatory protein EHD1 Is required for ocular lens development. Dev Biol 2015; 408:41-55. [PMID: 26455409 DOI: 10.1016/j.ydbio.2015.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 09/01/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022]
Abstract
The C-terminal Eps15 homology domain-containing (EHD) proteins play a key role in endocytic recycling, a fundamental cellular process that ensures the return of endocytosed membrane components and receptors back to the cell surface. To define the in vivo biological functions of EHD1, we have generated Ehd1 knockout mice and previously reported a requirement of EHD1 for spermatogenesis. Here, we show that approximately 56% of the Ehd1-null mice displayed gross ocular abnormalities, including anophthalmia, aphakia, microphthalmia and congenital cataracts. Histological characterization of ocular abnormalities showed pleiotropic defects that include a smaller or absent lens, persistence of lens stalk and hyaloid vasculature, and deformed optic cups. To test whether these profound ocular defects resulted from the loss of EHD1 in the lens or in non-lenticular tissues, we deleted the Ehd1 gene selectively in the presumptive lens ectoderm using Le-Cre. Conditional Ehd1 deletion in the lens resulted in developmental defects that included thin epithelial layers, small lenses and absence of corneal endothelium. Ehd1 deletion in the lens also resulted in reduced lens epithelial proliferation, survival and expression of junctional proteins E-cadherin and ZO-1. Finally, Le-Cre-mediated deletion of Ehd1 in the lens led to defects in corneal endothelial differentiation. Taken together, these data reveal a unique role for EHD1 in early lens development and suggest a previously unknown link between the endocytic recycling pathway and regulation of key developmental processes including proliferation, differentiation and morphogenesis.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Mark A Rainey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center Omaha, NE 68198-5900, USA.
| | - Bhopal C Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center Omaha, NE 68198-5870, USA.
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Murali R Kuracha
- Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA.
| | - Vimla Band
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA.
| | - Venkatesh Govindarajan
- Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA.
| | - Hamid Band
- Department of Genetics, Cell Biology & Anatomy, College of Medicine, University of Nebraska Medical Center, 985805 Nebraska Medical Center Omaha, NE 68198-5805, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, NE 68198-5950, USA; Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center Omaha, NE 68198-5900, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA.
| |
Collapse
|
43
|
Demonbreun AR, Biersmith BH, McNally EM. Membrane fusion in muscle development and repair. Semin Cell Dev Biol 2015; 45:48-56. [PMID: 26537430 PMCID: PMC4679555 DOI: 10.1016/j.semcdb.2015.10.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
Mature skeletal muscle forms from the fusion of skeletal muscle precursor cells, myoblasts. Myoblasts fuse to other myoblasts to generate multinucleate myotubes during myogenesis, and myoblasts also fuse to other myotubes during muscle growth and repair. Proteins within myoblasts and myotubes regulate complex processes such as elongation, migration, cell adherence, cytoskeletal reorganization, membrane coalescence, and ultimately fusion. Recent studies have identified cell surface proteins, intracellular proteins, and extracellular signaling molecules required for the proper fusion of muscle. Many proteins that actively participate in myoblast fusion also coordinate membrane repair. Here we will review mammalian membrane fusion with specific attention to proteins that mediate myoblast fusion and muscle repair.
Collapse
|
44
|
Matsudaira T, Niki T, Taguchi T, Arai H. Transport of the cholera toxin B-subunit from recycling endosomes to the Golgi requires clathrin and AP-1. J Cell Sci 2015; 128:3131-42. [PMID: 26136365 DOI: 10.1242/jcs.172171] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/22/2015] [Indexed: 11/20/2022] Open
Abstract
The retrograde pathway is defined by the transport of proteins and lipids from the plasma membrane through endosomes to the Golgi complex, and is essential for a variety of cellular activities. Recycling endosomes are important sorting stations for some retrograde cargo. SMAP2, a GTPase-activating protein (GAP) for Arf1 with a putative clathrin-binding domain, has previously been shown to participate in the retrograde transport of the cholera toxin B-subunit (CTxB) from recycling endosomes. Here, we found that clathrin, a vesicle coat protein, and clathrin adaptor protein complex 1 (AP-1) were present at recycling endosomes and were needed for the retrograde transport of CTxB from recycling endosomes to the Golgi, but not from the plasma membrane to recycling endosomes. SMAP2 immunoprecipitated clathrin and AP-1 through a putative clathrin-binding domain and a CALM-binding domain, and SMAP2 mutants that did not interact with clathrin or AP-1 could not localize to recycling endosomes. Moreover, knockdown of Arf1 suppressed the retrograde transport of CTxB from recycling endosomes to the Golgi. These findings suggest that retrograde transport is mediated by clathrin-coated vesicles from recycling endosomes and that the role of the coat proteins is in the recruitment of Arf GAP to transport vesicles.
Collapse
Affiliation(s)
- Tatsuyuki Matsudaira
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takahiro Niki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomohiko Taguchi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
45
|
Bahl K, Naslavsky N, Caplan S. Role of the EHD2 unstructured loop in dimerization, protein binding and subcellular localization. PLoS One 2015; 10:e0123710. [PMID: 25875965 PMCID: PMC4398442 DOI: 10.1371/journal.pone.0123710] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/05/2015] [Indexed: 11/20/2022] Open
Abstract
The C-terminal Eps 15 Homology Domain proteins (EHD1-4) play important roles in regulating endocytic trafficking. EHD2 is the only family member whose crystal structure has been solved, and it contains an unstructured loop consisting of two proline-phenylalanine (PF) motifs: KPFRKLNPF. In contrast, despite EHD2 having nearly 70% amino acid identity with its paralogs, EHD1, EHD3 and EHD4, the latter proteins contain a single KPF or RPF motif, but no NPF motif. In this study, we sought to define the precise role of each PF motif in EHD2’s homo-dimerization, binding with the protein partners, and subcellular localization. To test the role of the NPF motif, we generated an EHD2 NPF-to-NAF mutant to mimic the homologous sequences of EHD1 and EHD3. We demonstrated that this mutant lost both its ability to dimerize and bind to Syndapin2. However, it continued to localize primarily to the cytosolic face of the plasma membrane. On the other hand, EHD2 NPF-to-APA mutants displayed normal dimerization and Syndapin2 binding, but exhibited markedly increased nuclear localization and reduced association with the plasma membrane. We then hypothesized that the single PF motif of EHD1 (that aligns with the KPF of EHD2) might be responsible for both binding and localization functions of EHD1. Indeed, the EHD1 RPF motif was required for dimerization, interaction with MICAL-L1 and Syndapin2, as well as localization to tubular recycling endosomes. Moreover, recycling assays demonstrated that EHD1 RPF-to-APA was incapable of supporting normal receptor recycling. Overall, our data suggest that the EHD2 NPF phenylalanine residue is crucial for EHD2 localization to the plasma membrane, whereas the proline residue is essential for EHD2 dimerization and binding. These studies support the recently proposed model in which the EHD2 N-terminal region may regulate the availability of the unstructured loop for interactions with neighboring EHD2 dimers, thus promoting oligomerization.
Collapse
Affiliation(s)
- Kriti Bahl
- Department of Biochemistry and Molecular Biology, the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (SC); (NN)
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail: (SC); (NN)
| |
Collapse
|
46
|
Lee S, Uchida Y, Wang J, Matsudaira T, Nakagawa T, Kishimoto T, Mukai K, Inaba T, Kobayashi T, Molday RS, Taguchi T, Arai H. Transport through recycling endosomes requires EHD1 recruitment by a phosphatidylserine translocase. EMBO J 2015; 34:669-88. [PMID: 25595798 PMCID: PMC4365035 DOI: 10.15252/embj.201489703] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
P4-ATPases translocate aminophospholipids, such as phosphatidylserine (PS), to the cytosolic leaflet of membranes. PS is highly enriched in recycling endosomes (REs) and is essential for endosomal membrane traffic. Here, we show that PS flipping by an RE-localized P4-ATPase is required for the recruitment of the membrane fission protein EHD1. Depletion of ATP8A1 impaired the asymmetric transbilayer distribution of PS in REs, dissociated EHD1 from REs, and generated aberrant endosomal tubules that appear resistant to fission. EHD1 did not show membrane localization in cells defective in PS synthesis. ATP8A2, a tissue-specific ATP8A1 paralogue, is associated with a neurodegenerative disease (CAMRQ). ATP8A2, but not the disease-causative ATP8A2 mutant, rescued the endosomal defects in ATP8A1-depleted cells. Primary neurons from Atp8a2-/- mice showed a reduced level of transferrin receptors at the cell surface compared to Atp8a2+/+ mice. These findings demonstrate the role of P4-ATPase in membrane fission and give insight into the molecular basis of CAMRQ.
Collapse
Affiliation(s)
- Shoken Lee
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Yasunori Uchida
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Jiao Wang
- Departments of Biochemistry and Molecular Biology and Ophthalmology and Visual Sciences, Centre for Macular Research University of British Columbia, Vancouver, BC, Canada
| | - Tatsuyuki Matsudaira
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Takatoshi Nakagawa
- Department of Pharmacology, Osaka Medical College, Takatsuki-city Osaka, Japan
| | | | - Kojiro Mukai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Lipid Biology Laboratory, RIKEN, Wako-shi Saitama, Japan
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, Wako-shi Saitama, Japan
| | | | - Robert S Molday
- Departments of Biochemistry and Molecular Biology and Ophthalmology and Visual Sciences, Centre for Macular Research University of British Columbia, Vancouver, BC, Canada
| | - Tomohiko Taguchi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan Pathological Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences University of Tokyo, Tokyo, Japan
| |
Collapse
|
47
|
|
48
|
Zhang J, Naslavsky N, Caplan S. EHDs meet the retromer: Complex regulation of retrograde transport. CELLULAR LOGISTICS 2014. [PMID: 23181199 PMCID: PMC3498075 DOI: 10.4161/cl.20582] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Retrograde trafficking mediates the transport of endocytic membranes from endosomes to the trans-Golgi network (TGN). Dysregulation of these pathways can result in multiple ailments, including late-onset Alzheimer disease. One of the key retrograde transport regulators, the retromer complex, is tightly controlled by many factors, including the C-terminal Eps15 homology domain (EHD) proteins. This mini-review focuses on recent findings and discusses the regulation of the retromer complex by EHD proteins and the novel EHD1 interaction partner, Rabankyrin-5 (Rank-5).
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center; University of Nebraska Medical Center; Omaha, NE USA
| | | | | |
Collapse
|
49
|
SEC-10 and RAB-10 coordinate basolateral recycling of clathrin-independent cargo through endosomal tubules in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2014; 111:15432-7. [PMID: 25301900 DOI: 10.1073/pnas.1408327111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite the increasing number of regulatory proteins identified in clathrin-independent endocytic (CIE) pathways, our understanding of the exact functions of these proteins and the sequential manner in which they function remains limited. In this study, using the Caenorhabditis elegans intestine as a model, we observed a unique structure of interconnected endosomal tubules, which is required for the basolateral recycling of several CIE cargoes including hTAC, GLUT1, and DAF-4. SEC-10 is a subunit of the octameric protein complex exocyst. Depleting SEC-10 and several other exocyst components disrupted the endosomal tubules into various ring-like structures. An epistasis analysis further suggested that SEC-10 operates at the intermediate step between early endosomes and recycling endosomes. The endosomal tubules were also sensitive to inactivation of the Rab GTPase RAB-10 and disruption of microtubules. Taken together, our data suggest that SEC-10 coordinates with RAB-10 and microtubules to form the endosomal tubular network for efficient recycling of particular CIE cargoes.
Collapse
|
50
|
Mellado M, Cuartero Y, Brugada R, Verges M. Subcellular localisation of retromer in post-endocytic pathways of polarised Madin-Darby canine kidney cells. Biol Cell 2014; 106:377-93. [PMID: 25081925 DOI: 10.1111/boc.201400011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/28/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND INFORMATION Retromer is required for endosome-to-Golgi retrieval of the cation-independent mannose 6-phosphate receptor (CI-MPR), allowing delivery of hydrolases into lysosomes. It is constituted by a conserved heterotrimer formed by vacuolar protein sorting (Vps) gene products Vps26, Vps35 and Vps29, which is in charge of cargo selection, and a dimer of phosphoinositide-binding sorting nexins (SNXs), which has a structural role. Retromer has been implicated in sorting of additional cargo. Thus, retromer also promotes polymeric immunoglobulin A (pIgA) transcytosis by the pIgA receptor (pIgR) in polarised cells, and considerable evidence implicates retromer in controlling epithelial cell polarity. However, the precise localisation of retromer along the endocytic pathway of polarised cells has not been studied in detail. RESULTS Our biochemical analysis using rat liver endosome fractions suggests a distinct distribution pattern. Although subunits of the cargo-selective complex were enriched in early endosomes (EEs), levels of SNX2 were greater in sorting endosomes. We then immunolocalised the retromer subunits in polarised Madin-Darby canine kidney (MDCK) cells by confocal microscopy. An estimated 25% of total Vps26 and SNX2 localised to EEs, with negligible portions in recycling endosomes as well as in late endosomes and lysosomes. Although Vps26 was in structures of more heterogeneous size and shape than SNX2, these markedly overlapped. In consequence, the two retromer subcomplexes mostly colocalised. When we analysed retromer overlap with its cargo, we found that structures retromer and pIgA(+) are independent of those structures retromer and CI-MPR(+) . Remarkably, retromer localised preferentially at the transcytotic pathway. Pharmacological inhibition of phosphoinositide 3-kinase affected the co-distribution of retromer with pIgA and the CI-MPR, delaying pIgA progress to the apical surface. CONCLUSIONS In polarised MDCK cells, we found retromer associated with certain specialised EE-derived pathways. Our data imply that retromer is largely engaged in pIgA transcytosis in pIgR-expressing MDCK cells, as opposed to endosome-to-Golgi retrieval.
Collapse
Affiliation(s)
- Maravillas Mellado
- Laboratory of Epithelial Cell Biology, Príncipe Felipe Research Center (CIPF), Valencia, 46012, Spain
| | | | | | | |
Collapse
|