1
|
Zhu G, Zhang H, Xia M, Liu Y, Li M. EH domain-containing protein 2 (EHD2): Overview, biological function, and therapeutic potential. Cell Biochem Funct 2024; 42:e4016. [PMID: 38613224 DOI: 10.1002/cbf.4016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
EH domain-containing protein 2 (EHD2) is a member of the EHD protein family and is mainly located in the plasma membrane, but can also be found in the cytoplasm and endosomes. EHD2 is also a nuclear-cytoplasmic shuttle protein. After entering the cell nuclear, EHD2 acts as a corepressor of transcription to inhibit gene transcription. EHD2 regulates a series of biological processes. As a key regulator of endocytic transport, EHD2 is involved in the formation and maintenance of endosomal tubules and vesicles, which are critical for the intracellular transport of proteins and other substances. The N-terminal of EHD2 is attached to the cell membrane, while its C-terminal binds to the actin-binding protein. After binding, EHD2 connects with the actin cytoskeleton, forming the curvature of the membrane and promoting cell endocytosis. EHD2 is also associated with membrane protein trafficking and receptor signaling, as well as in glucose metabolism and lipid metabolism. In this review, we highlight the recent advances in the function of EHD2 in various cellular processes and its potential implications in human diseases such as cancer and metabolic disease. We also discussed the prospects for the future of EHD2. EHD2 has a broad prospect as a therapeutic target for a variety of diseases. Further research is needed to explore its mechanism, which could pave the way for the development of targeted treatments.
Collapse
Affiliation(s)
- Guoqiang Zhu
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hu Zhang
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Min Xia
- Hengyang Medical School, Institute of Clinical Medicine, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Hengyang Medical School, Cancer Research Institute, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yiqi Liu
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mingyong Li
- Department of Urology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Ji Y, Izadi-Seitz M, Landmann A, Schwintzer L, Qualmann B, Kessels MM. EHBP1 Is Critically Involved in the Dendritic Arbor Formation and Is Coupled to Factors Promoting Actin Filament Formation. J Neurosci 2024; 44:e0236232023. [PMID: 38129132 PMCID: PMC10860635 DOI: 10.1523/jneurosci.0236-23.2023] [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: 02/07/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The coordinated action of a plethora of factors is required for the organization and dynamics of membranous structures critically underlying the development and function of cells, organs, and organisms. The evolutionary acquisition of additional amino acid motifs allows for expansion and/or specification of protein functions. We identify a thus far unrecognized motif specific for chordata EHBP1 proteins and demonstrate that this motif is critically required for interaction with syndapin I, an F-BAR domain-containing, membrane-shaping protein predominantly expressed in neurons. Gain-of-function and loss-of-function studies in rat primary hippocampal neurons (of mixed sexes) unraveled that EHBP1 has an important role in neuromorphogenesis. Surprisingly, our analyses uncovered that this newly identified function of EHBP1 did not require the domain responsible for Rab GTPase binding but was strictly dependent on EHBP1's syndapin I binding interface and on the presence of syndapin I in the developing neurons. These findings were underscored by temporally and spatially remarkable overlapping dynamics of EHBP1 and syndapin I at nascent dendritic branch sites. In addition, rescue experiments demonstrated the necessity of two additional EHBP1 domains for dendritic arborization, the C2 and CH domains. Importantly, the additionally uncovered critical involvement of the actin nucleator Cobl in EHBP1 functions suggested that not only static association with F-actin via EHBP1's CH domain is important for dendritic arbor formation but also actin nucleation. Syndapin interactions organize ternary protein complexes composed of EHBP1, syndapin I, and Cobl, and our functional data show that only together these factors give rise to proper cell shape during neuronal development.
Collapse
Affiliation(s)
- Yuanyuan Ji
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Maryam Izadi-Seitz
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Annemarie Landmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Lukas Schwintzer
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
3
|
Rodriguez-Polanco WR, Norris A, Velasco AB, Gleason AM, Grant BD. Syndapin and GTPase RAP-1 control endocytic recycling via RHO-1 and non-muscle myosin II. Curr Biol 2023; 33:4844-4856.e5. [PMID: 37832552 PMCID: PMC10841897 DOI: 10.1016/j.cub.2023.09.051] [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: 04/14/2023] [Revised: 08/07/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023]
Abstract
After endocytosis, many plasma membrane components are recycled via membrane tubules that emerge from early endosomes to form recycling endosomes, eventually leading to their return to the plasma membrane. We previously showed that Syndapin/PACSIN-family protein SDPN-1 is required in vivo for basolateral endocytic recycling in the C. elegans intestine. Here, we document an interaction between the SDPN-1 SH3 domain and a target sequence in PXF-1/PDZ-GEF1/RAPGEF2, a known exchange factor for Rap-GTPases. We found that endogenous mutations engineered into the SDPN-1 SH3 domain, or its binding site in the PXF-1 protein, interfere with recycling in vivo, as does the loss of the PXF-1 target RAP-1. In some contexts, Rap-GTPases negatively regulate RhoA activity, suggesting a potential for Syndapin to regulate RhoA. Our results indicate that in the C. elegans intestine, RHO-1/RhoA is enriched on SDPN-1- and RAP-1-positive endosomes, and the loss of SDPN-1 or RAP-1 elevates RHO-1(GTP) levels on intestinal endosomes. Furthermore, we found that depletion of RHO-1 suppressed sdpn-1 mutant recycling defects, indicating that control of RHO-1 activity is a key mechanism by which SDPN-1 acts to promote endocytic recycling. RHO-1/RhoA is well known for controlling actomyosin contraction cycles, although little is known about the effects of non-muscle myosin II on endosomes. Our analysis found that non-muscle myosin II is enriched on SDPN-1-positive endosomes, with two non-muscle myosin II heavy-chain isoforms acting in apparent opposition. Depletion of nmy-2 inhibited recycling like sdpn-1 mutants, whereas depletion of nmy-1 suppressed sdpn-1 mutant recycling defects, indicating that actomyosin contractility controls recycling endosome function.
Collapse
Affiliation(s)
| | - Anne Norris
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Agustin B Velasco
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Adenrele M Gleason
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA; Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA; Rutgers Center for Lipid Research, Rutgers, the State University of New Jersey, New Brunswick, NJ 08901-8521, USA.
| |
Collapse
|
4
|
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
|
5
|
Izadi M, Wolf D, Seemann E, Ori A, Schwintzer L, Steiniger F, Kessels MM, Qualmann B. Membrane shapers from two distinct superfamilies cooperate in the development of neuronal morphology. J Cell Biol 2023; 222:e202211032. [PMID: 37318382 PMCID: PMC10274853 DOI: 10.1083/jcb.202211032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/27/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Membrane-shaping proteins are driving forces behind establishment of proper cell morphology and function. Yet, their reported structural and in vitro properties are noticeably inconsistent with many physiological membrane topology requirements. We demonstrate that dendritic arborization of neurons is powered by physically coordinated shaping mechanisms elicited by members of two distinct classes of membrane shapers: the F-BAR protein syndapin I and the N-Ank superfamily protein ankycorbin. Strikingly, membrane-tubulating activities by syndapin I, which would be detrimental during dendritic branching, were suppressed by ankycorbin. Ankycorbin's integration into syndapin I-decorated membrane surfaces instead promoted curvatures and topologies reflecting those observed physiologically. In line with the functional importance of this mechanism, ankycorbin- and syndapin I-mediated functions in dendritic arborization mutually depend on each other and on a surprisingly specific interface mediating complex formation of the two membrane shapers. These striking results uncovered cooperative and interdependent functions of members of two fundamentally different membrane shaper superfamilies as a previously unknown, pivotal principle in neuronal shape development.
Collapse
Affiliation(s)
- Maryam Izadi
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - David Wolf
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - Alessandro Ori
- Leibniz Institute on Aging—Fritz Lipmann Institute, Jena, Germany
| | - Lukas Schwintzer
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - Frank Steiniger
- Electron Microscopy Center, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - Michael Manfred Kessels
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
6
|
Rodriguez-Polanco WR, Norris A, Velasco AB, Gleason AM, Grant BD. Syndapin Regulates the RAP-1 GTPase to Control Endocytic Recycling via RHO-1 and Non-Muscle Myosin II. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530328. [PMID: 36909525 PMCID: PMC10002613 DOI: 10.1101/2023.02.27.530328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
After endocytosis, many plasma membrane components are recycled via narrow-diameter membrane tubules that emerge from early endosomes to form recycling endosomes, eventually leading to their return to the plasma membrane. We previously showed that the F-BAR and SH3 domain Syndapin/PACSIN-family protein SDPN-1 is required in vivo for basolateral endocytic recycling in the C. elegans intestine. Here we sought to determine the significance of a predicted interaction between the SDPN-1 SH3 domain and a target sequence in PXF-1/PDZ-GEF1/RAPGEF2, a known exchange factor for Rap-GTPases. We found that endogenous mutations we engineered into the SDPN-1 SH3 domain, or its binding site in the PXF-1 protein, interfere with recycling in vivo , as does loss of the PXF-1 target RAP-1. Rap-GTPases have been shown in several contexts to negatively regulate RhoA activity. Our results show that RHO-1/RhoA is enriched on SDPN-1 and RAP-1 positive endosomes in the C. elegans intestine, and loss of SDPN-1 or RAP-1 elevates RHO-1(GTP) levels on intestinal endosomes. Furthermore, we found that depletion of RHO-1 suppressed sdpn-1 mutant recycling defects, indicating that control of RHO-1 activity is a key mechanism by which SDPN-1 acts to promote endocytic recycling. RHO-1/RhoA is well-known for controlling actomyosin contraction cycles, although little is known of non-muscle myosin II on endosomes. Our analysis found that non-muscle myosin II is enriched on SDPN-1 positive endosomes, with two non-muscle myosin II heavy chain isoforms acting in apparent opposition. Depletion of nmy-2 inhibited recycling like sdpn-1 mutants, while depletion of nmy-1 suppressed sdpn-1 mutant recycling defects, indicating actomyosin contractility in controlling recycling endosome function.
Collapse
|
7
|
Cryo-electron tomography reveals structural insights into the membrane remodeling mode of dynamin-like EHD filaments. Nat Commun 2022; 13:7641. [PMID: 36496453 PMCID: PMC9741607 DOI: 10.1038/s41467-022-35164-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Eps15-homology domain containing proteins (EHDs) are eukaryotic, dynamin-related ATPases involved in cellular membrane trafficking. They oligomerize on membranes into filaments that induce membrane tubulation. While EHD crystal structures in open and closed conformations were previously reported, little structural information is available for the membrane-bound oligomeric form. Consequently, mechanistic insights into the membrane remodeling mechanism have remained sparse. Here, by using cryo-electron tomography and subtomogram averaging, we determined structures of nucleotide-bound EHD4 filaments on membrane tubes of various diameters at an average resolution of 7.6 Å. Assembly of EHD4 is mediated via interfaces in the G-domain and the helical domain. The oligomerized EHD4 structure resembles the closed conformation, where the tips of the helical domains protrude into the membrane. The variation in filament geometry and tube radius suggests a spontaneous filament curvature of approximately 1/70 nm-1. Combining the available structural and functional data, we suggest a model for EHD-mediated membrane remodeling.
Collapse
|
8
|
Tröger J, Seemann E, Heintzmann R, Kessels MM, Qualmann B. Spinal Cord Synaptic Plasticity by GlyRβ Release from Receptor Fields and Syndapin I-Dependent Uptake. J Neurosci 2022; 42:6706-6723. [PMID: 35879097 PMCID: PMC9436020 DOI: 10.1523/jneurosci.2060-21.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 06/23/2022] [Accepted: 07/13/2022] [Indexed: 11/21/2022] Open
Abstract
Glycine receptor-mediated inhibitory neurotransmission is key for spinal cord function. Recent observations suggested that by largely elusive mechanisms also glycinergic synapses display synaptic plasticity. We imaged receptor fields at ultrahigh-resolution at freeze-fractured membranes, tracked surface and internalized glycine receptors (GlyR), and studied differential regulations of GlyRβ interactions with the scaffold protein gephyrin and the F-BAR domain protein syndapin I and thereby reveal key principles of this process. S403 phosphorylation of GlyRβ, known to be triggered by synaptic signaling, caused a decoupling from gephyrin scaffolds but simultaneously promoted association of syndapin I with GlyRβ. In line, kainate treatments used to trigger rearrangements of glycine receptors in murine syndapin I KO spinal cords (mixed sex) showed even more severe receptor field fragmentation than already observed in untreated syndapin I KO spinal cords. Syndapin I deficiency furthermore resulted in more dispersed receptors and increased receptor mobility, also pointing out an important contribution of syndapin I to the organization of GlyRβ fields. Strikingly, syndapin I KO also led to a complete disruption of kainate-induced GlyRβ internalization. Accompanying quantitative ultrahigh-resolution studies in dissociated spinal cord neurons proved that the defects in GlyR internalization observed in syndapin I KO spinal cords are neuron-intrinsic defects caused by syndapin I deficiency. Together, our results unveiled important mechanisms organizing and altering glycine receptor fields during both steady state and particularly also as a consequence of kainate-induced synaptic rearrangement - principles organizing and fine-tuning synaptic efficacy and plasticity of glycinergic synapses in the spinal cord.SIGNIFICANCE STATEMENT Initial observations suggested that also glycinergic synapses, key for spinal cord and brainstem functions, may display some form of synaptic plasticity. Imaging receptor fields at ultrahigh-resolution at freeze-fractured membranes, tracking surface and internalized glycine receptors (GlyR) and studying regulations of GlyRβ interactions, we here reveal key principles of these kainate-inducible adaptations. A switch from gephyrin-mediated receptor scaffolding to syndapin I-mediated GlyRβ scaffolding and internalization allows for modulating synaptic receptor availability. In line, kainate-induced GlyRβ internalization was completely disrupted and GlyRβ receptor fields were distorted by syndapin I KO. These results unveiled important mechanisms during both steady-state and kainate-induced alterations of synaptic GlyR fields, principles underlying synaptic efficacy and plasticity of synapses in the spinal cord.
Collapse
Affiliation(s)
- Jessica Tröger
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, Jena 07743, Germany
| | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, Jena 07743, Germany
| | - Rainer Heintzmann
- Leibniz Institute of Photonic Technology, Jena 07745, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University, Jena 07745, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, Jena 07743, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich Schiller University Jena, Jena 07743, Germany
| |
Collapse
|
9
|
Morgan J, Yarwood R, Starborg T, Yan G, Lowe M. Pacsin2 is required for endocytosis in the zebrafish pronephric tubule. Biol Open 2022; 11:275521. [PMID: 35616009 PMCID: PMC9235069 DOI: 10.1242/bio.059150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022] Open
Abstract
Endocytosis mediates the cellular uptake of numerous molecules from the extracellular space and is a fundamentally important process. In the renal proximal tubule, the scavenger receptor megalin and its co-receptor cubilin mediate endocytosis of low molecular weight proteins from the renal filtrate. However, the extent to which megalin endocytosis relies on different components of the trafficking machinery remains relatively poorly defined in vivo. In this study, we identify a functional requirement for the F-BAR protein pacsin2 in endocytosis in the renal proximal tubule of zebrafish larvae. Pacsin2 is expressed throughout development and in all zebrafish tissues, similar to the mammalian orthologue. Within renal tubular epithelial cells, pacsin2 is enriched at the apical pole where it is localised to endocytic structures. Loss of pacsin2 results in reduced endocytosis within the proximal tubule, which is accompanied by a reduction in the abundance of megalin and endocytic organelles. Our results indicate that pacsin2 is required for efficient endocytosis in the proximal tubule, where it likely cooperates with other trafficking machinery to maintain endocytic uptake and recycling of megalin. Summary: We identify a role for the F-BAR protein pacsin2 in endocytosis in the renal tubule of zebrafish larvae.
Collapse
Affiliation(s)
- Joseph Morgan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Rebecca Yarwood
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Tobias Starborg
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Guanhua Yan
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
10
|
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: 2.0] [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
|
11
|
Dumont V, Lehtonen S. PACSIN proteins in vivo: Roles in development and physiology. Acta Physiol (Oxf) 2022; 234:e13783. [PMID: 34990060 PMCID: PMC9285741 DOI: 10.1111/apha.13783] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/15/2021] [Accepted: 01/01/2022] [Indexed: 12/22/2022]
Abstract
Protein kinase C and casein kinase substrate in neurons (PACSINs), or syndapins (synaptic dynamin‐associated proteins), are a family of proteins involved in the regulation of cell cytoskeleton, intracellular trafficking and signalling. Over the last twenty years, PACSINs have been mostly studied in the in vitro and ex vivo settings, and only in the last decade reports on their function in vivo have emerged. We first summarize the identification, structure and cellular functions of PACSINs, and then focus on the relevance of PACSINs in vivo. During development in various model organisms, PACSINs participate in diverse processes, such as neural crest cell development, gastrulation, laterality development and neuromuscular junction formation. In mouse, PACSIN2 regulates angiogenesis during retinal development and in human, PACSIN2 associates with monosomy and embryonic implantation. In adulthood, PACSIN1 has been extensively studied in the brain and shown to regulate neuromorphogenesis, receptor trafficking and synaptic plasticity. Several genetic studies suggest a role for PACSIN1 in the development of schizophrenia, which is also supported by the phenotype of mice depleted of PACSIN1. PACSIN2 plays an essential role in the maintenance of intestinal homeostasis and participates in kidney repair processes after injury. PACSIN3 is abundant in muscle tissue and necessary for caveolar biogenesis to create membrane reservoirs, thus controlling muscle function, and has been linked to certain genetic muscular disorders. The above examples illustrate the importance of PACSINs in diverse physiological or tissue repair processes in various organs, and associations to diseases when their functions are disturbed.
Collapse
Affiliation(s)
- Vincent Dumont
- Department of Pathology and Research Program for Clinical and Molecular Metabolism Faculty of Medicine University of Helsinki Helsinki Finland
| | - Sanna Lehtonen
- Department of Pathology and Research Program for Clinical and Molecular Metabolism Faculty of Medicine University of Helsinki Helsinki Finland
- Department of Pathology University of Helsinki Helsinki Finland
| |
Collapse
|
12
|
Ivanova D, Cousin MA. Synaptic Vesicle Recycling and the Endolysosomal System: A Reappraisal of Form and Function. Front Synaptic Neurosci 2022; 14:826098. [PMID: 35280702 PMCID: PMC8916035 DOI: 10.3389/fnsyn.2022.826098] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/03/2022] [Indexed: 12/15/2022] Open
Abstract
The endolysosomal system is present in all cell types. Within these cells, it performs a series of essential roles, such as trafficking and sorting of membrane cargo, intracellular signaling, control of metabolism and degradation. A specific compartment within central neurons, called the presynapse, mediates inter-neuronal communication via the fusion of neurotransmitter-containing synaptic vesicles (SVs). The localized recycling of SVs and their organization into functional pools is widely assumed to be a discrete mechanism, that only intersects with the endolysosomal system at specific points. However, evidence is emerging that molecules essential for endolysosomal function also have key roles within the SV life cycle, suggesting that they form a continuum rather than being isolated processes. In this review, we summarize the evidence for key endolysosomal molecules in SV recycling and propose an alternative model for membrane trafficking at the presynapse. This includes the hypotheses that endolysosomal intermediates represent specific functional SV pools, that sorting of cargo to SVs is mediated via the endolysosomal system and that manipulation of this process can result in both plastic changes to neurotransmitter release and pathophysiology via neurodegeneration.
Collapse
Affiliation(s)
- Daniela Ivanova
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Daniela Ivanova,
| | - Michael A. Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
- Michael A. Cousin,
| |
Collapse
|
13
|
Lysine acetylation regulates the interaction between proteins and membranes. Nat Commun 2021; 12:6466. [PMID: 34753925 PMCID: PMC8578602 DOI: 10.1038/s41467-021-26657-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/30/2021] [Indexed: 11/23/2022] Open
Abstract
Lysine acetylation regulates the function of soluble proteins in vivo, yet it remains largely unexplored whether lysine acetylation regulates membrane protein function. Here, we use bioinformatics, biophysical analysis of recombinant proteins, live-cell fluorescent imaging and genetic manipulation of Drosophila to explore lysine acetylation in peripheral membrane proteins. Analysis of 50 peripheral membrane proteins harboring BAR, PX, C2, or EHD membrane-binding domains reveals that lysine acetylation predominates in membrane-interaction regions. Acetylation and acetylation-mimicking mutations in three test proteins, amphiphysin, EHD2, and synaptotagmin1, strongly reduce membrane binding affinity, attenuate membrane remodeling in vitro and alter subcellular localization. This effect is likely due to the loss of positive charge, which weakens interactions with negatively charged membranes. In Drosophila, acetylation-mimicking mutations of amphiphysin cause severe disruption of T-tubule organization and yield a flightless phenotype. Our data provide mechanistic insights into how lysine acetylation regulates membrane protein function, potentially impacting a plethora of membrane-related processes. Lysine acetylation regulates the function of soluble proteins in vivo, yet it remains largely unexplored whether lysine acetylation regulates the function of membrane proteins. Here, the authors map lysine acetylation predominantly in membrane-interaction regions in peripheral membrane proteins and show with three candidate proteins how lysine acetylation is a regulator of membrane protein function.
Collapse
|
14
|
Wunderley L, Zhang L, Yarwood R, Qin W, Lowe M, Woodman P. Endosomal recycling tubule scission and integrin recycling involve the membrane curvature-supporting protein LITAF. J Cell Sci 2021; 134:jcs258549. [PMID: 34342350 PMCID: PMC8353527 DOI: 10.1242/jcs.258549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/16/2021] [Indexed: 11/20/2022] Open
Abstract
Recycling to the cell surface requires the scission of tubular membrane intermediates emanating from endosomes. Here, we identify the monotopic membrane protein LPS-induced TNF-activating factor (LITAF) and the related protein cell death involved p53 target 1 (CDIP1) as novel membrane curvature proteins that contribute to recycling tubule scission. Recombinant LITAF supports high membrane curvature, shown by its ability to reduce proteoliposome size. The membrane domains of LITAF and CDIP1 partition strongly into ∼50 nm diameter tubules labelled with the recycling markers Pacsin2, ARF6 and SNX1, and the recycling cargoes MHC class I and CD59. Partitioning of LITAF into tubules is impaired by mutations linked to Charcot Marie Tooth disease type 1C. Meanwhile, co-depletion of LITAF and CDIP1 results in the expansion of tubular recycling compartments and stabilised Rab11 tubules, pointing to a function for LITAF and CDIP1 in membrane scission. Consistent with this, co-depletion of LITAF and CDIP1 impairs integrin recycling and cell migration.
Collapse
Affiliation(s)
| | | | | | | | | | - Philip Woodman
- Faculty of Biology Medicine and Health, Manchester Academic and Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
15
|
Izadi M, Seemann E, Schlobinski D, Schwintzer L, Qualmann B, Kessels MM. Functional interdependence of the actin nucleator Cobl and Cobl-like in dendritic arbor development. eLife 2021; 10:67718. [PMID: 34264190 PMCID: PMC8282341 DOI: 10.7554/elife.67718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Local actin filament formation is indispensable for development of the dendritic arbor of neurons. We show that, surprisingly, the action of single actin filament-promoting factors was insufficient for powering dendritogenesis. Instead, this required the actin nucleator Cobl and its only evolutionary distant ancestor Cobl-like acting interdependently. This coordination between Cobl-like and Cobl was achieved by physical linkage by syndapins. Syndapin I formed nanodomains at convex plasma membrane areas at the base of protrusive structures and interacted with three motifs in Cobl-like, one of which was Ca2+/calmodulin-regulated. Consistently, syndapin I, Cobl-like’s newly identified N terminal calmodulin-binding site and the single Ca2+/calmodulin-responsive syndapin-binding motif all were critical for Cobl-like’s functions. In dendritic arbor development, local Ca2+/CaM-controlled actin dynamics thus relies on regulated and physically coordinated interactions of different F-actin formation-promoting factors and only together they have the power to bring about the sophisticated neuronal morphologies required for neuronal network formation in mammals.
Collapse
Affiliation(s)
- Maryam Izadi
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Dirk Schlobinski
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Lukas Schwintzer
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
16
|
Xu JJ, Li HD, Du XS, Li JJ, Meng XM, Huang C, Li J. Role of the F-BAR Family Member PSTPIP2 in Autoinflammatory Diseases. Front Immunol 2021; 12:585412. [PMID: 34262554 PMCID: PMC8273435 DOI: 10.3389/fimmu.2021.585412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Proline-serine-threonine-phosphatase-interacting protein 2 (PSTPIP2) belongs to the Fes/CIP4 homology-Bin/Amphiphysin/Rvs (F-BAR) domain family. It exhibits lipid-binding, membrane deformation, and F-actin binding activity, suggesting broader roles at the membrane–cytoskeleton interface. PSTPIP2 is known to participate in macrophage activation, neutrophil migration, cytokine production, and osteoclast differentiation. In recent years, it has been observed to play important roles in innate immune diseases and autoinflammatory diseases (AIDs). Current research indicates that the protein tyrosine phosphatase PTP-PEST, Src homology domain-containing inositol 5’-phosphatase 1 (SHIP1), and C‐terminal Src kinase (CSK) can bind to PSTPIP2 and inhibit the development of AIDs. However, the mechanisms underlying the function of PSTPIP2 have not been fully elucidated. This article reviews the research progress and mechanisms of PSTPIP2 in AIDs. PSTPIP2 also provides a new therapeutic target for the treatment of AIDs.
Collapse
Affiliation(s)
- Jie-Jie Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Di Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Sa Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan-Juan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Sakane A, Yano TA, Uchihashi T, Horikawa K, Hara Y, Imoto I, Kurisu S, Yamada H, Takei K, Sasaki T. JRAB/MICAL-L2 undergoes liquid-liquid phase separation to form tubular recycling endosomes. Commun Biol 2021; 4:551. [PMID: 33976349 PMCID: PMC8113518 DOI: 10.1038/s42003-021-02080-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
Elongated tubular endosomes play essential roles in diverse cellular functions. Multiple molecules have been implicated in tubulation of recycling endosomes, but the mechanism of endosomal tubule biogenesis has remained unclear. In this study, we found that JRAB/MICAL-L2 induces endosomal tubulation via activated Rab8A. In association with Rab8A, JRAB/MICAL-L2 adopts its closed form, which functions in the tubulation of recycling endosomes. Moreover, JRAB/MICAL-L2 induces liquid–liquid phase separation, initiating the formation of tubular recycling endosomes upon overexpression. Between its N-terminal and C-terminal globular domains, JRAB/MICAL-L2 contains an intrinsically disordered region, which contributes to the formation of JRAB/MICAL-L2 condensates. Based on our findings, we propose that JRAB/MICAL-L2 plays two sequential roles in the biogenesis of tubular recycling endosomes: first, JRAB/MICAL-L2 organizes phase separation, and then the closed form of JRAB/MICAL-L2 formed by interaction with Rab8A promotes endosomal tubulation. Sakane et al. demonstrate that JRAB/MICAL-L2, an effector protein of Rab8 and Rab13, induces endosomal tubulation in HeLa cells depending on its closed conformation caused by an activated Rab8A. JRAB/MICAL-L2 undergoes liquid-liquid phase separation when overexpressed, which precedes its interaction with Rab8A, eventually leading to tubulation.
Collapse
Affiliation(s)
- Ayuko Sakane
- Department of Biochemistry, Tokushima University Graduate School of Medical Sciences, Tokushima, Japan. .,Department of Interdisciplinary Researches for Medicine and Photonics, Institute of Post-LED Photonics, Tokushima, Japan.
| | - Taka-Aki Yano
- Department of Post-LED Photonics Research, Institute of Post-LED Photonics, Tokushima, Japan
| | - Takayuki Uchihashi
- Department of Physics, Nagoya University, Nagoya, Japan.,Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuki Horikawa
- Department of Optical Imaging, Advanced Research Promotion Center, Tokushima University, Tokushima, Japan
| | - Yusuke Hara
- Department of Optical Imaging, Advanced Research Promotion Center, Tokushima University, Tokushima, Japan
| | - Issei Imoto
- Division of Molecular Genetics, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Genetics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shusaku Kurisu
- Department of Cell Biology, Tokushima University Graduate School of Medical Sciences, Tokushima, Japan
| | - Hiroshi Yamada
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kohji Takei
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Sasaki
- Department of Biochemistry, Tokushima University Graduate School of Medical Sciences, Tokushima, Japan.
| |
Collapse
|
18
|
A junctional PACSIN2/EHD4/MICAL-L1 complex coordinates VE-cadherin trafficking for endothelial migration and angiogenesis. Nat Commun 2021; 12:2610. [PMID: 33972531 PMCID: PMC8110786 DOI: 10.1038/s41467-021-22873-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Angiogenic sprouting relies on collective migration and coordinated rearrangements of endothelial leader and follower cells. VE-cadherin-based adherens junctions have emerged as key cell-cell contacts that transmit forces between cells and trigger signals during collective cell migration in angiogenesis. However, the underlying molecular mechanisms that govern these processes and their functional importance for vascular development still remain unknown. We previously showed that the F-BAR protein PACSIN2 is recruited to tensile asymmetric adherens junctions between leader and follower cells. Here we report that PACSIN2 mediates the formation of endothelial sprouts during angiogenesis by coordinating collective migration. We show that PACSIN2 recruits the trafficking regulators EHD4 and MICAL-L1 to the rear end of asymmetric adherens junctions to form a recycling endosome-like tubular structure. The junctional PACSIN2/EHD4/MICAL-L1 complex controls local VE-cadherin trafficking and thereby coordinates polarized endothelial migration and angiogenesis. Our findings reveal a molecular event at force-dependent asymmetric adherens junctions that occurs during the tug-of-war between endothelial leader and follower cells, and allows for junction-based guidance during collective migration in angiogenesis. Communication between endothelial leader and follower cells during collective cell migration is crucial for vascular development. Here, the authors show that PACSIN2 guides collective cell migration and angiogenesis by recruiting a protein trafficking complex to asymmetric cell-cell junctions, controlling local junction plasticity.
Collapse
|
19
|
Yan Y, Liu S, Hu C, Xie C, Zhao L, Wang S, Zhang W, Cheng Z, Gao J, Fu X, Yang Z, Wang X, Zhang J, Lin L, Shi A. RTKN-1/Rhotekin shields endosome-associated F-actin from disassembly to ensure endocytic recycling. J Cell Biol 2021; 220:211976. [PMID: 33844824 PMCID: PMC8047894 DOI: 10.1083/jcb.202007149] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/22/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cargo sorting and the subsequent membrane carrier formation require a properly organized endosomal actin network. To better understand the actin dynamics during endocytic recycling, we performed a genetic screen in C. elegans and identified RTKN-1/Rhotekin as a requisite to sustain endosome-associated actin integrity. Loss of RTKN-1 led to a prominent decrease in actin structures and basolateral recycling defects. Furthermore, we showed that the presence of RTKN-1 thwarts the actin disassembly competence of UNC-60A/cofilin. Consistently, in RTKN-1–deficient cells, UNC-60A knockdown replenished actin structures and alleviated the recycling defects. Notably, an intramolecular interaction within RTKN-1 could mediate the formation of oligomers. Overexpression of an RTKN-1 mutant form that lacks self-binding capacity failed to restore actin structures and recycling flow in rtkn-1 mutants. Finally, we demonstrated that SDPN-1/Syndapin acts to direct the recycling endosomal dwelling of RTKN-1 and promotes actin integrity there. Taken together, these findings consolidated the role of SDPN-1 in organizing the endosomal actin network architecture and introduced RTKN-1 as a novel regulatory protein involved in this process.
Collapse
Affiliation(s)
- Yanling Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuai Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Can Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaoyi Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Linyue Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shimin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenjuan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zihang Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinghu Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhenrong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianghong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Long Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anbing Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
20
|
Farmer T, Xie S, Naslavsky N, Stöckli J, James DE, Caplan S. Defining the protein and lipid constituents of tubular recycling endosomes. J Biol Chem 2021; 296:100190. [PMID: 33334886 PMCID: PMC7948492 DOI: 10.1074/jbc.ra120.015992] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
Once internalized, receptors reach the sorting endosome and are either targeted for degradation or recycled to the plasma membrane, a process mediated at least in part by tubular recycling endosomes (TREs). TREs may be efficient for sorting owing to the ratio of large surface membrane area to luminal volume; following receptor segregation, TRE fission likely releases receptor-laden tubules and vesicles for recycling. Despite the importance of TRE networks for recycling, these unique structures remain poorly understood, and unresolved questions relate to their lipid and protein composition and biogenesis. Our previous studies have depicted the endocytic protein MICAL-L1 as an essential TRE constituent, and newer studies show a similar localization for the GTP-binding protein Rab10. We demonstrate that TREs are enriched in both phosphatidic acid (PA) and phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2), supporting the idea of MICAL-L1 recruitment by PA and Rab10 recruitment via PI(4,5)P2. Using siRNA knock-down, we demonstrate that Rab10-marked TREs remain prominent in cells upon MICAL-L1 or Syndapin2 depletion. However, depletion of Rab10 or its interaction partner, EHBP1, led to loss of MICAL-L1-marked TREs. We next used phospholipase D inhibitors to decrease PA synthesis, acutely disrupt TREs, and enable monitoring of TRE regeneration after inhibitor washout. Rab10 depletion prevented TRE regeneration, whereas MICAL-L1 knock-down did not. It is surprising that EHBP1 depletion did not affect TRE regeneration under these conditions. Overall, our study supports a primary role for Rab10 and the requirement for PA and PI(4,5)P2 in TRE biogenesis and regeneration, with Rab10 likely linking the sorting endosome to motor proteins and the microtubule network.
Collapse
Affiliation(s)
- Trey Farmer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shuwei Xie
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
21
|
Broad Kinase Inhibition Mitigates Early Neuronal Dysfunction in Tauopathy. Int J Mol Sci 2021; 22:ijms22031186. [PMID: 33530349 PMCID: PMC7865413 DOI: 10.3390/ijms22031186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/26/2022] Open
Abstract
Tauopathies are a group of more than twenty known disorders that involve progressive neurodegeneration, cognitive decline and pathological tau accumulation. Current therapeutic strategies provide only limited, late-stage symptomatic treatment. This is partly due to lack of understanding of the molecular mechanisms linking tau and cellular dysfunction, especially during the early stages of disease progression. In this study, we treated early stage tau transgenic mice with a multi-target kinase inhibitor to identify novel substrates that contribute to cognitive impairment and exhibit therapeutic potential. Drug treatment significantly ameliorated brain atrophy and cognitive function as determined by behavioral testing and a sensitive imaging technique called manganese-enhanced magnetic resonance imaging (MEMRI) with quantitative R1 mapping. Surprisingly, these benefits occurred despite unchanged hyperphosphorylated tau levels. To elucidate the mechanism behind these improved cognitive outcomes, we performed quantitative proteomics to determine the altered protein network during this early stage in tauopathy and compare this model with the human Alzheimer’s disease (AD) proteome. We identified a cluster of preserved pathways shared with human tauopathy with striking potential for broad multi-target kinase intervention. We further report high confidence candidate proteins as novel therapeutically relevant targets for the treatment of tauopathy. Proteomics data are available via ProteomeXchange with identifier PXD023562.
Collapse
|
22
|
Wang L, Ma H, Huang P, Xie Y, Near D, Wang H, Xu J, Yang Y, Xu Y, Garbutt T, Zhou Y, Liu Z, Yin C, Bressan M, Taylor JM, Liu J, Qian L. Down-regulation of Beclin1 promotes direct cardiac reprogramming. Sci Transl Med 2020; 12:eaay7856. [PMID: 33087505 PMCID: PMC8188650 DOI: 10.1126/scitranslmed.aay7856] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 05/07/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
Abstract
Direct reprogramming of fibroblasts to alternative cell fates by forced expression of transcription factors offers a platform to explore fundamental molecular events governing cell fate identity. The discovery and study of induced cardiomyocytes (iCMs) not only provides alternative therapeutic strategies for heart disease but also sheds lights on basic biology underlying CM fate determination. The iCM field has primarily focused on early transcriptome and epigenome repatterning, whereas little is known about how reprogramming iCMs remodel, erase, and exit the initial fibroblast lineage to acquire final cell identity. Here, we show that autophagy-related 5 (Atg5)-dependent autophagy, an evolutionarily conserved self-digestion process, was induced and required for iCM reprogramming. Unexpectedly, the autophagic factor Beclin1 (Becn1) was found to suppress iCM induction in an autophagy-independent manner. Depletion of Becn1 resulted in improved iCM induction from both murine and human fibroblasts. In a mouse genetic model, Becn1 haploinsufficiency further enhanced reprogramming factor-mediated heart function recovery and scar size reduction after myocardial infarction. Mechanistically, loss of Becn1 up-regulated Lef1 and down-regulated Wnt inhibitors, leading to activation of the canonical Wnt/β-catenin signaling pathway. In addition, Becn1 physically interacts with other classical class III phosphatidylinositol 3-kinase (PI3K III) complex components, the knockdown of which phenocopied Becn1 depletion in cardiac reprogramming. Collectively, our study revealed an inductive role of Atg5-dependent autophagy as well as a previously unrecognized autophagy-independent inhibitory function of Becn1 in iCM reprogramming.
Collapse
Affiliation(s)
- Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Peisen Huang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David Near
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yangxi Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tiffany Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yang Zhou
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ziqing Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chaoying Yin
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael Bressan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
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: 7] [Impact Index Per Article: 1.8] [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
|
24
|
Tom EC, Mushtaq I, Mohapatra BC, Luan H, Bhat AM, Zutshi N, Chakraborty S, Islam N, Arya P, Bielecki TA, Iseka FM, Bhattacharyya S, Cypher LR, Goetz BT, Negi SK, Storck MD, Rana S, Barnekow A, Singh PK, Ying G, Guda C, Natarajan A, Band V, Band H. EHD1 and RUSC2 Control Basal Epidermal Growth Factor Receptor Cell Surface Expression and Recycling. Mol Cell Biol 2020; 40:e00434-19. [PMID: 31932478 PMCID: PMC7076251 DOI: 10.1128/mcb.00434-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/26/2019] [Accepted: 12/26/2019] [Indexed: 01/25/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is a prototype receptor tyrosine kinase and an oncoprotein in many solid tumors. Cell surface display of EGFR is essential for cellular responses to its ligands. While postactivation endocytic trafficking of EGFR has been well elucidated, little is known about mechanisms of basal/preactivation surface display of EGFR. Here, we identify a novel role of the endocytic regulator EHD1 and a potential EHD1 partner, RUSC2, in cell surface display of EGFR. EHD1 and RUSC2 colocalize with EGFR in vesicular/tubular structures and at the Golgi compartment. Inducible EHD1 knockdown reduced the cell surface EGFR expression with accumulation at the Golgi compartment, a phenotype rescued by exogenous EHD1. RUSC2 knockdown phenocopied the EHD1 depletion effects. EHD1 or RUSC2 depletion impaired the EGF-induced cell proliferation, demonstrating that the novel, EHD1- and RUSC2-dependent transport of unstimulated EGFR from the Golgi compartment to the cell surface that we describe is functionally important, with implications for physiologic and oncogenic roles of EGFR and targeted cancer therapies.
Collapse
Affiliation(s)
- Eric C Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bhopal C Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Aaqib M Bhat
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Neha Zutshi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sukanya Chakraborty
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Namista Islam
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Priyanka Arya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fany M Iseka
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Luke R Cypher
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Simarjeet K Negi
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Angelika Barnekow
- Department of Experimental Tumorbiology, Westfälische Wilhelms University Muenster, Muenster, Germany
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, 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, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Guoguang Ying
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, 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, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
25
|
Koch N, Koch D, Krueger S, Tröger J, Sabanov V, Ahmed T, McMillan LE, Wolf D, Montag D, Kessels MM, Balschun D, Qualmann B. Syndapin I Loss-of-Function in Mice Leads to Schizophrenia-Like Symptoms. Cereb Cortex 2020; 30:4306-4324. [DOI: 10.1093/cercor/bhaa013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Schizophrenia is associated with cognitive and behavioral dysfunctions thought to reflect imbalances in neurotransmission systems. Recent screenings suggested that lack of (functional) syndapin I (PACSIN1) may be linked to schizophrenia. We therefore studied syndapin I KO mice to address the suggested causal relationship to schizophrenia and to analyze associated molecular, cellular, and neurophysiological defects. Syndapin I knockout (KO) mice developed schizophrenia-related behaviors, such as hyperactivity, reduced anxiety, reduced response to social novelty, and an exaggerated novel object response and exhibited defects in dendritic arborization in the cortex. Neuromorphogenic deficits were also observed for a schizophrenia-associated syndapin I mutant in cultured neurons and coincided with a lack of syndapin I–mediated membrane recruitment of cytoskeletal effectors. Syndapin I KO furthermore caused glutamatergic hypofunctions. Syndapin I regulated both AMPAR and NMDAR availabilities at synapses during basal synaptic activity and during synaptic plasticity—particularly striking were a complete lack of long-term potentiation and defects in long-term depression in syndapin I KO mice. These synaptic plasticity defects coincided with alterations of postsynaptic actin dynamics, synaptic GluA1 clustering, and GluA1 mobility. Both GluA1 and GluA2 were not appropriately internalized. Summarized, syndapin I KO led to schizophrenia-like behavior, and our analyses uncovered associated molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Nicole Koch
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Dennis Koch
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Sarah Krueger
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Jessica Tröger
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Victor Sabanov
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Tariq Ahmed
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Laura E McMillan
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - David Wolf
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Dirk Montag
- Neurogenetics Lab, Leibniz Institute for Neurobiology, 39116 Magdeburg, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Detlef Balschun
- Laboratory of Biological Psychology, Brain & Cognition, University of Leuven, 3000 Leuven, Belgium
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital—Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
26
|
The role of membrane-shaping BAR domain proteins in caveolar invagination: from mechanistic insights to pathophysiological consequences. Biochem Soc Trans 2020; 48:137-146. [DOI: 10.1042/bst20190377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 01/25/2023]
Abstract
The formation of caveolae, bulb-shaped plasma membrane invaginations, requires the coordinated action of distinct lipid-interacting and -shaping proteins. The interdependence of caveolar structure and function has evoked substantial scientific interest given the association of human diseases with caveolar dysfunction. Model systems deficient of core components of caveolae, caveolins or cavins, did not allow for an explicit attribution of observed functional defects to the requirement of caveolar invagination as they lack both invaginated caveolae and caveolin proteins. Knockdown studies in cultured cells and recent knockout studies in mice identified an additional family of membrane-shaping proteins crucial for caveolar formation, syndapins (PACSINs) — BAR domain superfamily proteins characterized by crescent-shaped membrane binding interfaces recognizing and inducing distinct curved membrane topologies. Importantly, syndapin loss-of-function resulted exclusively in impairment of caveolar invagination without a reduction in caveolin or cavin at the plasma membrane, thereby allowing the specific role of the caveolar invagination to be unveiled. Muscle cells of syndapin III KO mice showed severe reductions of caveolae reminiscent of human caveolinopathies and were more vulnerable to membrane damage upon changes in membrane tensions. Consistent with the lack of syndapin III-dependent invaginated caveolae providing mechanoprotection by releasing membrane reservoirs through caveolar flattening, physical exercise of syndapin III KO mice resulted in pathological defects reminiscent of the clinical symptoms of human myopathies associated with caveolin 3 mutation suggesting that the ability of muscular caveolae to respond to mechanical forces is a key physiological process.
Collapse
|
27
|
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: 3.8] [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
|
28
|
Cheung G, Cousin MA. Synaptic vesicle generation from activity-dependent bulk endosomes requires a dephosphorylation-dependent dynamin-syndapin interaction. J Neurochem 2019; 151:570-583. [PMID: 31479508 PMCID: PMC6899846 DOI: 10.1111/jnc.14862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/24/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Abstract
Activity‐dependent bulk endocytosis generates synaptic vesicles (SVs) during intense neuronal activity via a two‐step process. First, bulk endosomes are formed direct from the plasma membrane from which SVs are then generated. SV generation from bulk endosomes requires the efflux of previously accumulated calcium and activation of the protein phosphatase calcineurin. However, it is still unknown how calcineurin mediates SV generation. We addressed this question using a series of acute interventions that decoupled the generation of SVs from bulk endosomes in rat primary neuronal culture. This was achieved by either disruption of protein–protein interactions via delivery of competitive peptides, or inhibition of enzyme activity by known inhibitors. SV generation was monitored using either a morphological horseradish peroxidase assay or an optical assay that monitors the replenishment of the reserve SV pool. We found that SV generation was inhibited by, (i) peptides that disrupt calcineurin interactions, (ii) an inhibitor of dynamin I GTPase activity and (iii) peptides that disrupt the phosphorylation‐dependent dynamin I–syndapin I interaction. Peptides that disrupted syndapin I interactions with eps15 homology domain‐containing proteins had no effect. This revealed that (i) calcineurin must be localized at bulk endosomes to mediate its effect, (ii) dynamin I GTPase activity is essential for SV fission and (iii) the calcineurin‐dependent interaction between dynamin I and syndapin I is essential for SV generation. We therefore propose that a calcineurin‐dependent dephosphorylation cascade that requires both dynamin I GTPase and syndapin I lipid‐deforming activity is essential for SV generation from bulk endosomes. ![]()
Collapse
Affiliation(s)
- Giselle Cheung
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
29
|
Engevik AC, Kaji I, Postema MM, Faust JJ, Meyer AR, Williams JA, Fitz GN, Tyska MJ, Wilson JM, Goldenring JR. Loss of myosin Vb promotes apical bulk endocytosis in neonatal enterocytes. J Cell Biol 2019; 218:3647-3662. [PMID: 31562230 PMCID: PMC6829668 DOI: 10.1083/jcb.201902063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/22/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022] Open
Abstract
In patients with inactivating mutations in myosin Vb (Myo5B), enterocytes show large inclusions lined by microvilli. The origin of inclusions in small-intestinal enterocytes in microvillus inclusion disease is currently unclear. We postulated that inclusions in Myo5b KO mouse enterocytes form through invagination of the apical brush border membrane. 70-kD FITC-dextran added apically to Myo5b KO intestinal explants accumulated in intracellular inclusions. Live imaging of Myo5b KO-derived enteroids confirmed the formation of inclusions from the apical membrane. Treatment of intestinal explants and enteroids with Dyngo resulted in accumulation of inclusions at the apical membrane. Inclusions in Myo5b KO enterocytes contained VAMP4 and Pacsin 2 (Syndapin 2). Myo5b;Pacsin 2 double-KO mice showed a significant decrease in inclusion formation. Our results suggest that apical bulk endocytosis in Myo5b KO enterocytes resembles activity-dependent bulk endocytosis, the primary mechanism for synaptic vesicle uptake during intense neuronal stimulation. Thus, apical bulk endocytosis mediates the formation of inclusions in neonatal Myo5b KO enterocytes.
Collapse
Affiliation(s)
- Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Izumi Kaji
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - James J Faust
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Anne R Meyer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Janice A Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, Bio5 Institute, University of Arizona, Tucson, AZ
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN .,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN.,The Nashville VA Medical Center, Nashville, TN
| |
Collapse
|
30
|
Insinna C, Lu Q, Teixeira I, Harned A, Semler EM, Stauffer J, Magidson V, Tiwari A, Kenworthy AK, Narayan K, Westlake CJ. Investigation of F-BAR domain PACSIN proteins uncovers membrane tubulation function in cilia assembly and transport. Nat Commun 2019; 10:428. [PMID: 30683896 PMCID: PMC6347608 DOI: 10.1038/s41467-018-08192-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/20/2018] [Indexed: 12/03/2022] Open
Abstract
The intracellular ciliogenesis pathway requires membrane trafficking, fusion, and reorganization. Here, we demonstrate in human cells and zebrafish that the F-BAR domain containing proteins PACSIN1 and -2 play an essential role in ciliogenesis, similar to their binding partner and membrane reorganizer EHD1. In mature cilia, PACSINs and EHDs are dynamically localized to the ciliary pocket membrane (CPM) and transported away from this structure on membrane tubules along with proteins that exit the cilium. PACSINs function early in ciliogenesis at the ciliary vesicle (CV) stage to promote mother centriole to basal body transition. Remarkably, we show that PACSIN1 and EHD1 assemble membrane t7ubules from the developing intracellular cilium that attach to the plasma membrane, creating an extracellular membrane channel (EMC) to the outside of the cell. Together, our work uncovers a function for F-BAR proteins and membrane tubulation in ciliogenesis and explains how the intracellular cilium emerges from the cell.
Collapse
Affiliation(s)
- Christine Insinna
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Quanlong Lu
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Isabella Teixeira
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Adam Harned
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21701, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Elizabeth M Semler
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Jim Stauffer
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Valentin Magidson
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21701, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Christopher J Westlake
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| |
Collapse
|
31
|
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: 4.8] [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
|
32
|
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: 1.5] [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
|
33
|
Cullen PJ, Steinberg F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat Rev Mol Cell Biol 2018; 19:679-696. [PMID: 30194414 DOI: 10.1038/s41580-018-0053-7] [Citation(s) in RCA: 334] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Newly endocytosed integral cell surface proteins are typically either directed for degradation or subjected to recycling back to the plasma membrane. The sorting of integral cell surface proteins, including signalling receptors, nutrient transporters, ion channels, adhesion molecules and polarity markers, within the endolysosomal network for recycling is increasingly recognized as an essential feature in regulating the complexities of physiology at the cell, tissue and organism levels. Historically, endocytic recycling has been regarded as a relatively passive process, where the majority of internalized integral proteins are recycled via a nonspecific sequence-independent 'bulk membrane flow' pathway. Recent work has increasingly challenged this view. The discovery of sequence-specific sorting motifs and the identification of cargo adaptors and associated coat complexes have begun to uncover the highly orchestrated nature of endosomal cargo recycling, thereby providing new insight into the function and (patho)physiology of this process.
Collapse
Affiliation(s)
- Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK.
| | - Florian Steinberg
- Center for Biological Systems Analysis, Albert Ludwigs Universitaet Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
34
|
Activity-dependent bulk endocytosis proteome reveals a key presynaptic role for the monomeric GTPase Rab11. Proc Natl Acad Sci U S A 2018; 115:E10177-E10186. [PMID: 30301801 PMCID: PMC6205440 DOI: 10.1073/pnas.1809189115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The maintenance of neurotransmission by synaptic vesicle (SV) recycling is critical to brain function. The dominant SV recycling mode during intense activity is activity-dependent bulk endocytosis (ADBE), suggesting it will perform a pivotal role in neurotransmission. However, the role of ADBE is still undetermined, due to the absence of identified molecules specific for this process. The determination of the bulk endosome proteome (a key ADBE organelle) revealed that it has a unique molecular signature and identified a role for Rab11 in presynaptic function. This work provides the molecular inventory of ADBE, a resource that will be of significant value to researchers wishing to modulate neurotransmission during intense neuronal activity in both health and disease. Activity-dependent bulk endocytosis (ADBE) is the dominant mode of synaptic vesicle endocytosis during high-frequency stimulation, suggesting it should play key roles in neurotransmission during periods of intense neuronal activity. However, efforts in elucidating the physiological role of ADBE have been hampered by the lack of identified molecules which are unique to this endocytosis mode. To address this, we performed proteomic analysis on purified bulk endosomes, which are a key organelle in ADBE. Bulk endosomes were enriched via two independent approaches, a classical subcellular fractionation method and isolation via magnetic nanoparticles. There was a 77% overlap in proteins identified via the two protocols, and these molecules formed the ADBE core proteome. Bioinformatic analysis revealed a strong enrichment in cell adhesion and cytoskeletal and signaling molecules, in addition to expected synaptic and trafficking proteins. Network analysis identified Rab GTPases as a central hub within the ADBE proteome. Subsequent investigation of a subset of these Rabs revealed that Rab11 both facilitated ADBE and accelerated clathrin-mediated endocytosis. These findings suggest that the ADBE proteome will provide a rich resource for the future study of presynaptic function, and identify Rab11 as a regulator of presynaptic function.
Collapse
|
35
|
Stoddard EG, Volk RF, Carson JP, Ljungberg CM, Murphree TA, Smith JN, Sadler NC, Shukla AK, Ansong C, Wright AT. Multifunctional Activity-Based Protein Profiling of the Developing Lung. J Proteome Res 2018; 17:2623-2634. [PMID: 29972024 DOI: 10.1021/acs.jproteome.8b00086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung diseases and disorders are a leading cause of death among infants. Many of these diseases and disorders are caused by premature birth and underdeveloped lungs. In addition to developmentally related disorders, the lungs are exposed to a variety of environmental contaminants and xenobiotics upon birth that can cause breathing issues and are progenitors of cancer. In order to gain a deeper understanding of the developing lung, we applied an activity-based chemoproteomics approach for the functional characterization of the xenometabolizing cytochrome P450 enzymes, active ATP and nucleotide binding enzymes, and serine hydrolases using a suite of activity-based probes (ABPs). We detected P450 activity primarily in the postnatal lung; using our ATP-ABP, we characterized a wide range of ATPases and other active nucleotide- and nucleic acid-binding enzymes involved in multiple facets of cellular metabolism throughout development. ATP-ABP targets include kinases, phosphatases, NAD- and FAD-dependent enzymes, RNA/DNA helicases, and others. The serine hydrolase-targeting probe detected changes in the activities of several proteases during the course of lung development, yielding insights into protein turnover at different stages of development. Select activity-based probe targets were then correlated with RNA in situ hybridization analyses of lung tissue sections.
Collapse
Affiliation(s)
- Ethan G Stoddard
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Regan F Volk
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - James P Carson
- Texas Advanced Computing Center , University of Texas at Austin , Austin , Texas 78758 , United States
| | - Cecilia M Ljungberg
- Department of Pediatrics, Baylor College of Medicine , Jan and Dan Duncan Neurological Research Center at Texas Children's Hospital , Houston , Texas 77030 , United States
| | - Taylor A Murphree
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Jordan N Smith
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Natalie C Sadler
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Anil K Shukla
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Charles Ansong
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States
| | - Aaron T Wright
- Biological Sciences Division , Pacific Northwest National Laboratory , Richland , Washington 99352 , United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering , Washington State University , Pullman , Washington 99163 , United States
| |
Collapse
|
36
|
Abstract
The p2b domain of Rous sarcoma virus (RSV) Gag and the p6 domain of HIV-1 Gag contain late assembly (L) domains that engage the ESCRT membrane fission machinery and are essential for virus release. We now show that the PPXY-type RSV L domain specifically recruits the BAR domain protein PACSIN2 into virus-like particles (VLP), in addition to the NEDD4-like ubiquitin ligase ITCH and ESCRT pathway components such as TSG101. PACSIN2, which has been implicated in the remodeling of cellular membranes and the actin cytoskeleton, is also recruited by HIV-1 p6 independent of its ability to engage the ESCRT factors TSG101 or ALIX. Moreover, PACSIN2 is robustly recruited by NEDD4-2s, a NEDD4-like ubiquitin ligase capable of rescuing HIV-1 budding defects. The NEDD4-2s-induced incorporation of PACSIN2 into VLP correlated with the formation of Gag-ubiquitin conjugates, indicating that PACSIN2 binds ubiquitin. Although PACSIN2 was not required for a single cycle of HIV-1 replication after infection with cell-free virus, HIV-1 spreading was nevertheless severely impaired in T cell lines and primary human peripheral blood mononuclear cells depleted of PACSIN2. HIV-1 spreading could be restored by reintroduction of wild-type PACSIN2, but not of a SH3 domain mutant unable to interact with the actin polymerization regulators WASP and N-WASP. Overall, our observations indicate that PACSIN2 promotes the cell-to-cell spreading of HIV-1 by connecting Gag to the actin cytoskeleton.
Collapse
|
37
|
Van Quickelberghe E, Martens A, Goeminne LJE, Clement L, van Loo G, Gevaert K. Identification of Immune-Responsive Gene 1 (IRG1) as a Target of A20. J Proteome Res 2018; 17:2182-2191. [DOI: 10.1021/acs.jproteome.8b00139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Arne Martens
- VIB-UGent Center
for Inflammation Research, B-9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | | | | | - Geert van Loo
- VIB-UGent Center
for Inflammation Research, B-9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center
for Medical Biotechnology, B-9000 Ghent, Belgium
| |
Collapse
|
38
|
Izadi M, Schlobinski D, Lahr M, Schwintzer L, Qualmann B, Kessels MM. Cobl-like promotes actin filament formation and dendritic branching using only a single WH2 domain. J Cell Biol 2017; 217:211-230. [PMID: 29233863 PMCID: PMC5748978 DOI: 10.1083/jcb.201704071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 09/13/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023] Open
Abstract
Local actin filament formation powers the development of the signal-receiving arbor of neurons. In this study, Izadi et al. demonstrate that Cobl-like, which bears only a single WH2 domain, mediates dendritic branching by coordinating with the F-actin–binding protein Abp1 in a Ca2+/CaM-controlled manner to control actin dynamics. Local actin filament formation powers the development of the signal-receiving arbor of neurons that underlies neuronal network formation. Yet, little is known about the molecules that drive these processes and may functionally connect them to the transient calcium pulses observed in restricted areas in the forming dendritic arbor. Here we demonstrate that Cordon-Bleu (Cobl)–like, an uncharacterized protein suggested to represent a very distantly related, evolutionary ancestor of the actin nucleator Cobl, despite having only a single G-actin–binding Wiskott–Aldrich syndrome protein Homology 2 (WH2) domain, massively promoted the formation of F-actin–rich membrane ruffles of COS-7 cells and of dendritic branches of neurons. Cobl-like hereby integrates WH2 domain functions with those of the F-actin–binding protein Abp1. Cobl-like–mediated dendritic branching is dependent on Abp1 as well as on Ca2+/calmodulin (CaM) signaling and CaM association. Calcium signaling leads to a promotion of complex formation with Cobl-like’s cofactor Abp1. Thus, Ca2+/CaM control of actin dynamics seems to be a much more broadly used principle in cell biology than previously thought.
Collapse
Affiliation(s)
- Maryam Izadi
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Dirk Schlobinski
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Maria Lahr
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Lukas Schwintzer
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
39
|
Iseka FM, Goetz BT, Mushtaq I, An W, Cypher LR, Bielecki TA, Tom EC, Arya P, Bhattacharyya S, Storck MD, Semerad CL, Talmadge JE, Mosley RL, Band V, Band H. Role of the EHD Family of Endocytic Recycling Regulators for TCR Recycling and T Cell Function. THE JOURNAL OF IMMUNOLOGY 2017; 200:483-499. [PMID: 29212907 DOI: 10.4049/jimmunol.1601793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/01/2017] [Indexed: 12/31/2022]
Abstract
T cells use the endocytic pathway for key cell biological functions, including receptor turnover and maintenance of the immunological synapse. Some of the established players include the Rab GTPases, the SNARE complex proteins, and others, which function together with EPS-15 homology domain-containing (EHD) proteins in non-T cell systems. To date, the role of the EHD protein family in T cell function remains unexplored. We generated conditional EHD1/3/4 knockout mice using CD4-Cre and crossed these with mice bearing a myelin oligodendrocyte glycoprotein-specific TCR transgene. We found that CD4+ T cells from these mice exhibited reduced Ag-driven proliferation and IL-2 secretion in vitro. In vivo, these mice exhibited reduced severity of experimental autoimmune encephalomyelitis. Further analyses showed that recycling of the TCR-CD3 complex was impaired, leading to increased lysosomal targeting and reduced surface levels on CD4+ T cells of EHD1/3/4 knockout mice. Our studies reveal a novel role of the EHD family of endocytic recycling regulatory proteins in TCR-mediated T cell functions.
Collapse
Affiliation(s)
- Fany M Iseka
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Luke R Cypher
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Eric C Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Priyanka Arya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Craig L Semerad
- Flow Cytometry Research Facility, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - R Lee Mosley
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198; .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
40
|
Seemann E, Sun M, Krueger S, Tröger J, Hou W, Haag N, Schüler S, Westermann M, Huebner CA, Romeike B, Kessels MM, Qualmann B. Deciphering caveolar functions by syndapin III KO-mediated impairment of caveolar invagination. eLife 2017; 6. [PMID: 29202928 PMCID: PMC5716666 DOI: 10.7554/elife.29854] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022] Open
Abstract
Several human diseases are associated with a lack of caveolae. Yet, the functions of caveolae and the molecular mechanisms critical for shaping them still are debated. We show that muscle cells of syndapin III KO mice show severe reductions of caveolae reminiscent of human caveolinopathies. Yet, different from other mouse models, the levels of the plasma membrane-associated caveolar coat proteins caveolin3 and cavin1 were both not reduced upon syndapin III KO. This allowed for dissecting bona fide caveolar functions from those supported by mere caveolin presence and also demonstrated that neither caveolin3 nor caveolin3 and cavin1 are sufficient to form caveolae. The membrane-shaping protein syndapin III is crucial for caveolar invagination and KO rendered the cells sensitive to membrane tensions. Consistent with this physiological role of caveolae in counterpoising membrane tensions, syndapin III KO skeletal muscles showed pathological parameters upon physical exercise that are also found in CAVEOLIN3 mutation-associated muscle diseases.
Collapse
Affiliation(s)
- Eric Seemann
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Minxuan Sun
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Sarah Krueger
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Jessica Tröger
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Wenya Hou
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Natja Haag
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Susann Schüler
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Christian A Huebner
- Institute for Human Genetics, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Bernd Romeike
- Institute of Pathology, Division of Neuropathology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Michael M Kessels
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Britta Qualmann
- Institute for Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
41
|
Semmler J, Kormann J, Srinivasan SP, Köster A, Sälzer D, Reppel M, Hescheler J, Plomann M, Nguemo F. Pacsin 2 is required for the maintenance of a normal cardiac function in the developing mouse heart. Pharmacol Res 2017; 128:200-210. [PMID: 29107716 DOI: 10.1016/j.phrs.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/26/2017] [Accepted: 10/15/2017] [Indexed: 11/27/2022]
Abstract
The Pacsin proteins (Pacsin 1, 2 and 3) play an important role in intracellular trafficking and thereby signal transduction in many cells types. This study was designed to examine the role of Pacsin 2 in cardiac development and function. We investigated the development and electrophysiological properties of Pacsin 2 knockout (P2KO) hearts and single cardiomyocytes isolated from 11.5 and 15.5days old fetal mice. Immunofluorescence experiments confirmed the lack of Pacsin 2 protein expression in P2KO cardiac myocytes in comparison to wildtype (WT). Western blotting demonstrates low expression levels of connexin 43 and T-box 3 proteins in P2KO compared to wildtype (WT). Electrophysiology measurements including online Multi-Electrode Array (MEA) based field potential (FP) recordings on isolated whole heart of P2KO mice showed a prolonged AV-conduction time. Patch clamp measurements of P2KO cardiomyocytes revealed differences in action potential (AP) parameters and decreased pacemaker funny channel (If), as well as L-type Ca2+ channel (ICaL), and sodium channel (INa). These findings demonstrate that Pacsin 2 is necessary for cardiac development and function in mouse embryos, which will enhance our knowledge to better understand the genesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Judith Semmler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Jan Kormann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | | | - Annette Köster
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Daniel Sälzer
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany; Department of Cardiology, University of Lübeck, Lübeck, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Markus Plomann
- Institute of Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
42
|
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
|
43
|
Structural insights into the activation mechanism of dynamin-like EHD ATPases. Proc Natl Acad Sci U S A 2017; 114:5629-5634. [PMID: 28228524 DOI: 10.1073/pnas.1614075114] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Eps15 (epidermal growth factor receptor pathway substrate 15)-homology domain containing proteins (EHDs) comprise a family of dynamin-related mechano-chemical ATPases involved in cellular membrane trafficking. Previous studies have revealed the structure of the EHD2 dimer, but the molecular mechanisms of membrane recruitment and assembly have remained obscure. Here, we determined the crystal structure of an amino-terminally truncated EHD4 dimer. Compared with the EHD2 structure, the helical domains are 50° rotated relative to the GTPase domain. Using electron paramagnetic spin resonance (EPR), we show that this rotation aligns the two membrane-binding regions in the helical domain toward the lipid bilayer, allowing membrane interaction. A loop rearrangement in GTPase domain creates a new interface for oligomer formation. Our results suggest that the EHD4 structure represents the active EHD conformation, whereas the EHD2 structure is autoinhibited, and reveal a complex series of domain rearrangements accompanying activation. A comparison with other peripheral membrane proteins elucidates common and specific features of this activation mechanism.
Collapse
|
44
|
Gleason AM, Nguyen KCQ, Hall DH, Grant BD. Syndapin/SDPN-1 is required for endocytic recycling and endosomal actin association in the C. elegans intestine. Mol Biol Cell 2016; 27:mbc.E16-02-0116. [PMID: 27630264 PMCID: PMC5170557 DOI: 10.1091/mbc.e16-02-0116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/18/2016] [Accepted: 09/08/2016] [Indexed: 11/11/2022] Open
Abstract
Syndapin/Pascin family F-BAR domain proteins bind directly to membrane lipids and are associated with actin dynamics at the plasma membrane. Previous reports have also implicated mammalian syndapin 2 in endosome function during receptor recycling, but precise analysis of a putative recycling function for syndapin in mammalian systems is difficult because of syndapin effects on the earlier step of endocytic uptake, and potential redundancy among the three separate genes that encode mammalian syndapin isoforms. Here we analyze the endocytic transport function of the only C. elegans syndapin, SDPN-1. We find that SDPN-1 is a resident protein of the early and basolateral recycling endosomes in the C. elegans intestinal epithelium, and sdpn-1 deletion mutants display phenotypes indicating a block in basolateral recycling transport. sdpn-1 mutants accumulate abnormal endosomes positive for early endosome and recycling endosome markers that are normally separate, and such endosomes accumulate high levels of basolateral recycling cargo. Furthermore, we observed strong colocalization of endosomal SDPN-1 with the F-actin biosensor Lifeact, and found that loss of SDPN-1 greatly reduced Lifeact accumulation on early endosomes. Taken together our results provide strong evidence for an in vivo function of syndapin in endocytic recycling, and suggest that syndapin promotes transport via endosomal fission.
Collapse
Affiliation(s)
- Adenrele M Gleason
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Ken C Q Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
45
|
PACSIN1 regulates the dynamics of AMPA receptor trafficking. Sci Rep 2016; 6:31070. [PMID: 27488904 PMCID: PMC4973260 DOI: 10.1038/srep31070] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 07/14/2016] [Indexed: 01/29/2023] Open
Abstract
Dynamic trafficking of AMPA receptors (AMPARs) into and out of synapses plays an important role in synaptic plasticity. We previously reported that the protein kinase C and casein kinase II substrate in neurons (PACSIN) forms a complex with AMPARs through its interaction with the protein interacting with C-kinase 1 (PICK1) to regulate NMDA receptor (NMDAR)-induced AMPAR endocytosis and cerebellar long-term depression. However, the molecular mechanism by which PACSIN regulates the dynamics of AMPAR trafficking remains unclear. Using a pH-sensitive green fluorescent protein, pHluorin, tagged to the extracellular domain of the GluA2 subunit of AMPARs, we demonstrate dual roles for PACSIN1 in controlling the internalization and recycling of GluA2 after NMDAR activation. Structure and function analysis reveals a requirement for the PACSIN1 F-BAR and SH3 domains in controlling these NMDAR-dependent processes. Interestingly, the variable region, which binds to PICK1, is not essential for NMDAR-dependent GluA2 internalization and is required only for the correct recycling of AMPARs. These results indicate that PACSIN is a versatile membrane deformation protein that links the endocytic and recycling machineries essential for dynamic AMPAR trafficking in neurons.
Collapse
|
46
|
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: 6] [Impact Index Per Article: 0.8] [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
|
47
|
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: 2.3] [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
|
48
|
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: 3.6] [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
|
49
|
Hou W, Izadi M, Nemitz S, Haag N, Kessels MM, Qualmann B. The Actin Nucleator Cobl Is Controlled by Calcium and Calmodulin. PLoS Biol 2015; 13:e1002233. [PMID: 26334624 PMCID: PMC4559358 DOI: 10.1371/journal.pbio.1002233] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 07/23/2015] [Indexed: 01/10/2023] Open
Abstract
Actin nucleation triggers the formation of new actin filaments and has the power to shape cells but requires tight control in order to bring about proper morphologies. The regulation of the members of the novel class of WASP Homology 2 (WH2) domain-based actin nucleators, however, thus far has largely remained elusive. Our study reveals signal cascades and mechanisms regulating Cordon-Bleu (Cobl). Cobl plays some, albeit not fully understood, role in early arborization of neurons and nucleates actin by a mechanism that requires a combination of all three of its actin monomer–binding WH2 domains. Our experiments reveal that Cobl is regulated by Ca2+ and multiple, direct associations of the Ca2+ sensor Calmodulin (CaM). Overexpression analyses and rescue experiments of Cobl loss-of-function phenotypes with Cobl mutants in primary neurons and in tissue slices demonstrated the importance of CaM binding for Cobl’s functions. Cobl-induced dendritic branch initiation was preceded by Ca2+ signals and coincided with local F-actin and CaM accumulations. CaM inhibitor studies showed that Cobl-mediated branching is strictly dependent on CaM activity. Mechanistic studies revealed that Ca2+/CaM modulates Cobl’s actin binding properties and furthermore promotes Cobl’s previously identified interactions with the membrane-shaping F-BAR protein syndapin I, which accumulated with Cobl at nascent dendritic protrusion sites. The findings of our study demonstrate a direct regulation of an actin nucleator by Ca2+/CaM and reveal that the Ca2+/CaM-controlled molecular mechanisms we discovered are crucial for Cobl’s cellular functions. By unveiling the means of Cobl regulation and the mechanisms, by which Ca2+/CaM signals directly converge on a cellular effector promoting actin filament formation, our work furthermore sheds light on how local Ca2+ signals steer and power branch initiation during early arborization of nerve cells—a key process in neuronal network formation. The calcium sensor calmodulin directly regulates the actin filament-promoting factor Cobl to help shape the complex architecture of neurons underlying neuronal network formation. The organization and the formation of new actin filaments by polymerization of actin monomers has the power to shape cells. The rate-limiting step in actin polymerization is “nucleation”—a process during which the first actin monomers are assembled with the help of actin nucleators. This nucleation step requires tight temporal and spatial control in order to achieve proper cell morphologies. Here, we analyse signaling cascades and mechanisms regulating the actin nucleator Cobl, which is crucial for the formation of dendritic arbors of nerve cells—a key process in neuronal network formation. We show that the calcium (Ca2+)-binding signaling component calmodulin (CaM) binds to Cobl and regulates its functions. Using 3-D time-lapse analyses of developing neurons, we visualized how Cobl works. We observed local accumulation of CaM, Cobl, actin, and syndapin I—a membrane-shaping protein—at dendritic branch initiation sites. We find that Ca2+/CaM modulates Cobl’s actin-binding properties and promotes its interactions with syndapin I, which then serves as a membrane anchor for Cobl. In summary, we i) show a direct regulation of the actin nucleator Cobl by Ca2+/CaM, ii) demonstrate that the molecular mechanisms we discovered are crucial for shaping nerve cells, and iii) underscore how local Ca2+ signals steer and power branch initiation during early arborization of neurons.
Collapse
Affiliation(s)
- Wenya Hou
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Maryam Izadi
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Sabine Nemitz
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Natja Haag
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Michael M. Kessels
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
- * E-mail: (BQ); (MMK)
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
- * E-mail: (BQ); (MMK)
| |
Collapse
|
50
|
Koles K, Messelaar EM, Feiger Z, Yu CJ, Frank CA, Rodal AA. The EHD protein Past1 controls postsynaptic membrane elaboration and synaptic function. Mol Biol Cell 2015. [PMID: 26202464 PMCID: PMC4569317 DOI: 10.1091/mbc.e15-02-0093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The conserved C-terminal EHD protein Past1 is required for postsynaptic membrane remodeling and synaptic transmission at the Drosophila neuromuscular junction. Past1 activity defines distinct synaptic microdomains of the BAR-domain proteins Syndapin and Amphiphysin, suggesting a new mechanism for elaboration of the postsynaptic membrane reticulum. Membranes form elaborate structures that are highly tailored to their specialized cellular functions, yet the mechanisms by which these structures are shaped remain poorly understood. Here, we show that the conserved membrane-remodeling C-terminal Eps15 Homology Domain (EHD) protein Past1 is required for the normal assembly of the subsynaptic muscle membrane reticulum (SSR) at the Drosophila melanogaster larval neuromuscular junction (NMJ). past1 mutants exhibit altered NMJ morphology, decreased synaptic transmission, reduced glutamate receptor levels, and a deficit in synaptic homeostasis. The membrane-remodeling proteins Amphiphysin and Syndapin colocalize with Past1 in distinct SSR subdomains and collapse into Amphiphysin-dependent membrane nodules in the SSR of past1 mutants. Our results suggest a mechanism by which the coordinated actions of multiple lipid-binding proteins lead to the elaboration of increasing layers of the SSR and uncover new roles for an EHD protein at synapses.
Collapse
Affiliation(s)
- Kate Koles
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02453
| | - Emily M Messelaar
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02453
| | - Zachary Feiger
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02453
| | - Crystal J Yu
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02453
| | - C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242
| | - Avital A Rodal
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02453
| |
Collapse
|