1
|
Wang J, Barr MM, Wehman AM. Extracellular vesicles. Genetics 2024; 227:iyae088. [PMID: 38884207 PMCID: PMC11304975 DOI: 10.1093/genetics/iyae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Extracellular vesicles (EVs) encompass a diverse array of membrane-bound organelles released outside cells in response to developmental and physiological cell needs. EVs play important roles in remodeling the shape and content of differentiating cells and can rescue damaged cells from toxic or dysfunctional content. EVs can send signals and transfer metabolites between tissues and organisms to regulate development, respond to stress or tissue damage, or alter mating behaviors. While many EV functions have been uncovered by characterizing ex vivo EVs isolated from body fluids and cultured cells, research using the nematode Caenorhabditis elegans has provided insights into the in vivo functions, biogenesis, and uptake pathways. The C. elegans EV field has also developed methods to analyze endogenous EVs within the organismal context of development and adult physiology in free-living, behaving animals. In this review, we summarize major themes that have emerged for C. elegans EVs and their relevance to human health and disease. We also highlight the diversity of biogenesis mechanisms, locations, and functions of worm EVs and discuss open questions and unexplored topics tenable in C. elegans, given the nematode model is ideal for light and electron microscopy, genetic screens, genome engineering, and high-throughput omics.
Collapse
Affiliation(s)
- Juan Wang
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Maureen M Barr
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Ann M Wehman
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
2
|
Song JZ, Feng YH, Sergevnina V, Zhu J, Li H, Xie Z. Assessing the Presence of Phosphoinositides on Autophagosomal Membrane in Yeast by Live Cell Imaging. Microorganisms 2024; 12:1458. [PMID: 39065227 PMCID: PMC11279164 DOI: 10.3390/microorganisms12071458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The formation of autophagosomes mediating the sequestration of cytoplasmic materials is the central step of autophagy. Several phosphoinositides, which are signaling molecules on the membrane, are involved in autophagy. However, it is not always clear whether these phosphoinositides act directly at the site of autophagosome formation, or indirectly via the regulation of other steps or pathways. To address this question, we used a set of phosphoinositide probes to systematically examine their potential presence on autophagosomal membranes in yeast (Saccharomyces cerevisiae). We verified the specificity of these probes using mutant cells deficient in the production of the corresponding phosphoinositides. We then examined starved yeast cells co-expressing a phosphoinositide probe together with an autophagosomal membrane marker, 2Katushka2S-Atg8. Our data revealed that PtdIns(4,5)P2 and PtdIns(3,5)P2 were mainly present on the plasma membrane and vacuolar membrane, respectively. We observed only occasional co-localization between the PtdIns(4)P probe and Atg8, some of which may represent the transient passage of a PtdIns(4)P-containing structure near the autophagosomal membrane. In contrast, substantial colocalization of the PtdIns(3)P probe with Atg8 was observed. Taken together, our data indicate that only PtdIns(3)P is present in a substantial amount on the autophagosomal membrane. For other phosphoinositides involved in autophagy, either their presence on the autophagosomal membrane is very transient, or they act on other cellular membranes to regulate autophagy.
Collapse
Affiliation(s)
| | | | | | | | - Hui Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiping Xie
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
3
|
Muth LT, Van Bogaert INA. Let it stick: Strategies and applications for intracellular plasma membrane targeting of proteins in Saccharomyces cerevisiae. Yeast 2024; 41:315-329. [PMID: 38444057 DOI: 10.1002/yea.3933] [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: 07/06/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/07/2024] Open
Abstract
Lipid binding domains and protein lipidations are essential features to recruit proteins to intracellular membranes, enabling them to function at specific sites within the cell. Membrane association can also be exploited to answer fundamental and applied research questions, from obtaining insights into the understanding of lipid metabolism to employing them for metabolic engineering to redirect fluxes. This review presents a broad catalog of membrane binding strategies focusing on the plasma membrane of Saccharomyces cerevisiae. Both lipid binding domains (pleckstrin homology, discoidin-type C2, kinase associated-1, basic-rich and bacterial phosphoinositide-binding domains) and co- and post-translational lipidations (prenylation, myristoylation and palmitoylation) are introduced as tools to target the plasma membrane. To provide a toolset of membrane targeting modules, respective candidates that facilitate plasma membrane targeting are showcased including their in vitro and in vivo properties. The relevance and versatility of plasma membrane targeting modules are further highlighted by presenting a selected set of use cases.
Collapse
Affiliation(s)
- Liv Teresa Muth
- Department of Biotechnology, Centre for Synthetic Biology, Ghent University, Ghent, Belgium
| | | |
Collapse
|
4
|
Saha S, Krishnan H, Raghu P. IMPA1 dependent regulation of phosphatidylinositol 4,5-bisphosphate and calcium signalling by lithium. Life Sci Alliance 2024; 7:e202302425. [PMID: 38056909 PMCID: PMC10700560 DOI: 10.26508/lsa.202302425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
Lithium (Li) is widely used as a mood stabilizer to treat bipolar affective disorder. However, the molecular targets of Li that underpin its therapeutic effect remain unresolved. Inositol monophosphatase (IMPA1) is an enzyme involved in phosphatidylinositol 4,5-bisphosphate (PIP2) resynthesis after PLC signaling. In vitro, Li inhibits IMPA1, but the relevance of this inhibition within neural cells remains unknown. Here, we report that treatment with therapeutic concentrations of Li reduces receptor-activated calcium release from intracellular stores and delays PIP2 resynthesis. These effects of Li are abrogated in IMPA1 deleted cells. We also observed that in human forebrain cortical neurons, treatment with Li reduced neuronal excitability and calcium signals. After Li treatment of human cortical neurons, transcriptome analyses revealed down-regulation of signaling by glutamate, a key excitatory neurotransmitter in the human brain. Collectively, our findings suggest that inhibition of IMPA1 by Li reduces receptor-activated PLC signaling and neuronal excitability.
Collapse
Affiliation(s)
- Sankhanil Saha
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| | - Harini Krishnan
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| |
Collapse
|
5
|
Ubeysinghe S, Kankanamge D, Thotamune W, Wijayaratna D, Mohan TM, Karunarathne A. Spatiotemporal Optical Control of Gαq-PLCβ Interactions. ACS Synth Biol 2024; 13:242-258. [PMID: 38092428 DOI: 10.1021/acssynbio.3c00490] [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] [Indexed: 12/26/2023]
Abstract
Cells experience time-varying and spatially heterogeneous chemokine signals in vivo, activating cell surface proteins including G protein-coupled receptors (GPCRs). The Gαq pathway activation by GPCRs is a major signaling axis with broad physiological and pathological significance. Compared with other Gα members, GαqGTP activates many crucial effectors, including PLCβ (Phospholipase Cβ) and Rho GEFs (Rho guanine nucleotide exchange factors). PLCβ regulates many key processes, such as hematopoiesis, synaptogenesis, and cell cycle, and is therefore implicated in terminal-debilitating diseases, including cancer, epilepsy, Huntington's Disease, and Alzheimer's Disease. However, due to a lack of genetic and pharmacological tools, examining how the dynamic regulation of PLCβ signaling controls cellular physiology has been difficult. Since activated PLCβ induces several abrupt cellular changes, including cell morphology, examining how the other pathways downstream of Gq-GPCRs contribute to the overall signaling has also been difficult. Here we show the engineering, validation, and application of a highly selective and efficient optogenetic inhibitor (Opto-dHTH) to completely disrupt GαqGTP-PLCβ interactions reversibly in user-defined cellular-subcellular regions on optical command. Using this newly gained PLCβ signaling control, our data indicate that the molecular competition between RhoGEFs and PLCβ for GαqGTP determines the potency of Gq-GPCR-governed directional cell migration.
Collapse
Affiliation(s)
- Sithurandi Ubeysinghe
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Dinesh Kankanamge
- Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Waruna Thotamune
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Dhanushan Wijayaratna
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Thomas M Mohan
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Ajith Karunarathne
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| |
Collapse
|
6
|
Mondal S, Sarvari G, Boehr DD. Picornavirus 3C Proteins Intervene in Host Cell Processes through Proteolysis and Interactions with RNA. Viruses 2023; 15:2413. [PMID: 38140654 PMCID: PMC10747604 DOI: 10.3390/v15122413] [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: 10/15/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The Picornaviridae family comprises a large group of non-enveloped viruses with enormous impact on human and animal health. The picornaviral genome contains one open reading frame encoding a single polyprotein that can be processed by viral proteases. The picornaviral 3C proteases share similar three-dimensional structures and play a significant role in the viral life cycle and virus-host interactions. Picornaviral 3C proteins also have conserved RNA-binding activities that contribute to the assembly of the viral RNA replication complex. The 3C protease is important for regulating the host cell response through the cleavage of critical host cell proteins, acting to selectively 'hijack' host factors involved in gene expression, promoting picornavirus replication, and inactivating key factors in innate immunity signaling pathways. The protease and RNA-binding activities of 3C are involved in viral polyprotein processing and the initiation of viral RNA synthesis. Most importantly, 3C modifies critical molecules in host organelles and maintains virus infection by subtly subverting host cell death through the blocking of transcription, translation, and nucleocytoplasmic trafficking to modulate cell physiology for viral replication. Here, we discuss the molecular mechanisms through which 3C mediates physiological processes involved in promoting virus infection, replication, and release.
Collapse
Affiliation(s)
| | | | - David D. Boehr
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
7
|
Chiu TY, Lo CH, Lin YH, Lai YD, Lin SS, Fang YT, Huang WS, Huang SY, Tsai PY, Yang FH, Chong WM, Wu YC, Tsai HC, Liu YW, Hsu CL, Liao JC, Wang WJ. INPP5E regulates CD3ζ enrichment at the immune synapse by phosphoinositide distribution control. Commun Biol 2023; 6:911. [PMID: 37670137 PMCID: PMC10480498 DOI: 10.1038/s42003-023-05269-0] [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: 08/17/2022] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
The immune synapse, a highly organized structure formed at the interface between T lymphocytes and antigen-presenting cells (APCs), is essential for T cell activation and the adaptive immune response. It has been shown that this interface shares similarities with the primary cilium, a sensory organelle in eukaryotic cells, although the roles of ciliary proteins on the immune synapse remain elusive. Here, we find that inositol polyphosphate-5-phosphatase E (INPP5E), a cilium-enriched protein responsible for regulating phosphoinositide localization, is enriched at the immune synapse in Jurkat T-cells during superantigen-mediated conjugation or antibody-mediated crosslinking of TCR complexes, and forms a complex with CD3ζ, ZAP-70, and Lck. Silencing INPP5E in Jurkat T-cells impairs the polarized distribution of CD3ζ at the immune synapse and correlates with a failure of PI(4,5)P2 clearance at the center of the synapse. Moreover, INPP5E silencing decreases proximal TCR signaling, including phosphorylation of CD3ζ and ZAP-70, and ultimately attenuates IL-2 secretion. Our results suggest that INPP5E is a new player in phosphoinositide manipulation at the synapse, controlling the TCR signaling cascade.
Collapse
Grants
- National Science and Technology Council, Taiwan, NSTC 110-2326-B-A49A-503-MY3, 111-2628-B-A49A-016, and 112-2628-B-A49-009-MY3
- National Health Research Institutes (NHRI-EX109-10610BC) National Taiwan University and Academia Sinica Innovative Joint Program (109L104303)
- National Science and Technology Council, Taiwan, NSTC 109-2628-B-010-016 Cancer Progression Research Center NYCU, from the Higher Education Sprout Project by MOE
- National Science and Technology Council, Taiwan, NSTC 107-2313-B-001-009 National Science and Technology Council, Taiwan, NSTC 108-2313-B-001-003 National Taiwan University and Academia Sinica Innovative Joint Program Grant (NTU-SINICA- 108L104303)
Collapse
Affiliation(s)
- Tzu-Yuan Chiu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan
- The Scripps Research Institute, La Jolla, 92037, USA
| | - Chien-Hui Lo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Yi-Hsuan Lin
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Yun-Di Lai
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Shan-Shan Lin
- Institute of Molecular Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Ya-Tian Fang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan
| | - Wei-Syun Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Shen-Yan Huang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Pei-Yuan Tsai
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Fu-Hua Yang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan
| | - Weng Man Chong
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan
| | - Yi-Chieh Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Hsing-Chen Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100233, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Jung-Chi Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan.
- Syncell Inc., Taipei, 115202, Taiwan.
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan.
| |
Collapse
|
8
|
Ubeysinghe S, Kankanamge D, Thotamune W, Wijayaratna D, Mohan TM, Karunarathne A. Spatiotemporal optical control of Gαq-PLCβ interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552801. [PMID: 37609229 PMCID: PMC10441412 DOI: 10.1101/2023.08.10.552801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Cells experience time-varying and spatially heterogeneous chemokine signals in vivo, activating cell surface proteins, including G protein-coupled receptors (GPCRs). The Gαq pathway activation by GPCRs is a major signaling axis with a broad physiological and pathological significance. Compared to other Gα members, GαqGTP activates many crucial effectors, including PLCβ (Phospholipase Cβ) and Rho GEFs (Rho guanine nucleotide exchange factors). PLCβ regulates many key processes, such as hematopoiesis, synaptogenesis, and cell cycle, and is therefore implicated in terminal - debilitating diseases, including cancer, epilepsy, Huntington's Disease, and Alzheimer's Disease. However, due to a lack of genetic and pharmacological tools, examining how the dynamic regulation of PLCβ signaling controls cellular physiology has been difficult. Since activated PLCβ induces several abrupt cellular changes, including cell morphology, examining how the other pathways downstream of Gq-GPCRs contribute to the overall signaling has also been difficult. Here we show the engineering, validation, and application of a highly selective and efficient optogenetic inhibitor (Opto-dHTH) to completely disrupt GαqGTP-PLCβ interactions reversibly in user-defined cellular-subcellular regions on optical command. Using this newly gained PLCβ signaling control, our data indicate that the molecular competition between RhoGEFs and PLCβ for GαqGTP determines the potency of Gq-GPCR-governed directional cell migration.
Collapse
|
9
|
Thallmair V, Schultz L, Evers S, Jolie T, Goecke C, Leitner MG, Thallmair S, Oliver D. Localization of the tubby domain, a PI(4,5)P2 biosensor, to E-Syt3-rich endoplasmic reticulum-plasma membrane junctions. J Cell Sci 2023; 136:jcs260848. [PMID: 37401342 PMCID: PMC10445746 DOI: 10.1242/jcs.260848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/26/2023] [Indexed: 07/05/2023] Open
Abstract
The phospholipid phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2] acts as a signaling lipid at the plasma membrane (PM) with pleiotropic regulatory actions on multiple cellular processes. Signaling specificity might result from spatiotemporal compartmentalization of the lipid and from combinatorial binding of PI(4,5)P2 effector proteins to additional membrane components. Here, we analyzed the spatial distribution of tubbyCT, a paradigmatic PI(4,5)P2-binding domain, in live mammalian cells by total internal reflection fluorescence (TIRF) microscopy and molecular dynamics simulations. We found that unlike other well-characterized PI(4,5)P2 recognition domains, tubbyCT segregates into distinct domains within the PM. TubbyCT enrichment occurred at contact sites between PM and endoplasmic reticulum (ER) (i.e. at ER-PM junctions) as shown by colocalization with ER-PM markers. Localization to these sites was mediated in a combinatorial manner by binding to PI(4,5)P2 and by interaction with a cytosolic domain of extended synaptotagmin 3 (E-Syt3), but not other E-Syt isoforms. Selective localization to these structures suggests that tubbyCT is a novel selective reporter for a ER-PM junctional pool of PI(4,5)P2. Finally, we found that association with ER-PM junctions is a conserved feature of tubby-like proteins (TULPs), suggesting an as-yet-unknown function of TULPs.
Collapse
Affiliation(s)
- Veronika Thallmair
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps UniversityMarburg, 35037 Marburg, Germany
| | - Lea Schultz
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Saskia Evers
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Theresa Jolie
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Christian Goecke
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Michael G. Leitner
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH&Co.KG, Birkendorfer Str. 65, 88400 Biberach an der Riß, Germany
| | - Sebastian Thallmair
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Groningen Biomolecular Sciences and Biotechnology Institute and The Zernike Institute for Advanced Material, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps UniversityMarburg, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, 35032 Marburg, Germany
| |
Collapse
|
10
|
Le TPH, Nguyen NTT, Le DDT, Anwar MA, Lee SY. Lipid kinase PIP5Kα contributes to Hippo pathway activation via interaction with Merlin and by mediating plasma membrane targeting of LATS1. Cell Commun Signal 2023; 21:149. [PMID: 37337213 DOI: 10.1186/s12964-023-01161-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND The Hippo pathway plays a critical role in controlled cell proliferation. The tumor suppressor Merlin and large tumor suppressor kinase 1 (LATS1) mediate activation of Hippo pathway, consequently inhibiting the primary effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). Phosphatidylinositol 4,5-bisphosphate (PIP2), a lipid present in the plasma membrane (PM), binds to and activates Merlin. Phosphatidylinositol 4-phosphate 5-kinase α (PIP5Kα) is an enzyme responsible for PIP2 production. However, the functional role of PIP5Kα in regulation of Merlin and LATS1 under Hippo signaling conditions remains unclear. METHODS PIP5Kα, Merlin, or LATS1 knockout or knockdown cells and transfected cells with them were used. LATS1, YAP, and TAZ activities were measured using biochemical methods and PIP2 levels were evaluated using cell imaging. Low/high cell density and serum starvation/stimulation conditions were tested. Colocalization of PIP5Kα and PIP2 with Merlin and LATS1, and their protein interactions were examined using transfection, confocal imaging, immunoprecipitation, western blotting, and/or pull-down experiments. Colony formation and adipocyte differentiation assays were performed. RESULTS We found that PIP5Kα induced LATS1 activation and YAP/TAZ inhibition in a kinase activity-dependent manner. Consistent with these findings, PIP5Kα suppressed cell proliferation and enhanced adipocyte differentiation of mesenchymal stem cells. Moreover, PIP5Kα protein stability and PIP2 levels were elevated at high cell density compared with those at low cell density, and both PIP2 and YAP phosphorylation levels initially declined, then recovered upon serum stimulation. Under these conditions, YAP/TAZ activity was aberrantly regulated by PIP5Kα deficiency. Mechanistically, either Merlin deficiency or LATS1 deficiency abrogated PIP5Kα-mediated YAP/TAZ inactivation. Additionally, the catalytic domain of PIP5Kα directly interacted with the band 4.1/ezrin/radixin/moesin domain of Merlin, and this interaction reinforced interaction of Merlin with LATS1. In accordance with these findings, PIP5Kα and PIP2 colocalized with Merlin and LATS1 in the PM. In PIP5Kα-deficient cells, Merlin colocalization with PIP2 was reduced, and LATS1 solubility increased. CONCLUSIONS Collectively, our results support that PIP5Kα serves as an activator of the Hippo pathway through interaction and colocalization with Merlin, which promotes PIP2-dependent Merlin activation and induces local recruitment of LATS1 to the PIP2-rich PM and its activation, thereby negatively regulating YAP/TAZ activity. Video Abstract.
Collapse
Affiliation(s)
- Truc Phan Hoang Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Nga Thi Thanh Nguyen
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Duong Duy Thai Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea
| | - Muhammad Ayaz Anwar
- Department of Applied Chemistry, Kyung Hee University International Campus, Yongin, Gyeonggi, 17104, Republic of Korea
| | - Sang Yoon Lee
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea.
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi, 16499, Republic of Korea.
| |
Collapse
|
11
|
Raut P, Obeng B, Waters H, Zimmerberg J, Gosse JA, Hess ST. Phosphatidylinositol 4,5-Bisphosphate Mediates the Co-Distribution of Influenza A Hemagglutinin and Matrix Protein M1 at the Plasma Membrane. Viruses 2022; 14:v14112509. [PMID: 36423118 PMCID: PMC9698905 DOI: 10.3390/v14112509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The fully assembled influenza A virus (IAV) has on its surface the highest density of a single membrane protein found in nature-the glycoprotein hemagglutinin (HA) that mediates viral binding, entry, and assembly. HA clusters at the plasma membrane of infected cells, and the HA density (number of molecules per unit area) of these clusters correlates with the infectivity of the virus. Dense HA clusters are considered to mark the assembly site and ultimately lead to the budding of infectious IAV. The mechanism of spontaneous HA clustering, which occurs with or without other viral components, has not been elucidated. Using super-resolution fluorescence photoactivation localization microscopy (FPALM), we have previously shown that these HA clusters are interdependent on phosphatidylinositol 4,5-biphosphate (PIP2). Here, we show that the IAV matrix protein M1 co-clusters with PIP2, visualized using the pleckstrin homology domain. We find that cetylpyridinium chloride (CPC), which is a positively charged quaternary ammonium compound known for its antibacterial and antiviral properties at millimolar concentrations, disrupts M1 clustering and M1-PIP2 co-clustering at micromolar concentrations well below the critical micelle concentration (CMC). CPC also disrupts the co-clustering of M1 with HA at the plasma membrane, suggesting the role of host cell PIP2 clusters as scaffolds for gathering and concentrating M1 and HA to achieve their unusually high cluster densities in the IAV envelope.
Collapse
Affiliation(s)
- Prakash Raut
- Department of Physics and Astronomy, University of Maine, Orono, ME 04469-5709, USA
| | - Bright Obeng
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469-5735, USA
| | - Hang Waters
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Joshua Zimmerberg
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1855, USA
| | - Julie A. Gosse
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469-5735, USA
| | - Samuel T. Hess
- Department of Physics and Astronomy, University of Maine, Orono, ME 04469-5709, USA
- Correspondence:
| |
Collapse
|
12
|
Thallmair V, Schultz L, Zhao W, Marrink SJ, Oliver D, Thallmair S. Two cooperative binding sites sensitize PI(4,5)P 2 recognition by the tubby domain. SCIENCE ADVANCES 2022; 8:eabp9471. [PMID: 36070381 PMCID: PMC9451155 DOI: 10.1126/sciadv.abp9471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/20/2022] [Indexed: 05/22/2023]
Abstract
Phosphoinositides (PIs) are lipid signaling molecules that operate by recruiting proteins to cellular membranes via PI recognition domains. The dominant PI of the plasma membrane is phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. One of only two PI(4,5)P2 recognition domains characterized in detail is the tubby domain. It is essential for targeting proteins into cilia involving reversible membrane association. However, the PI(4,5)P2 binding properties of tubby domains have remained enigmatic. Here, we used coarse-grained molecular dynamics simulations to explore PI(4,5)P2 binding by the prototypic tubby domain. The comparatively low PI(4,5)P2 affinity of the previously described canonical binding site is underpinned in a cooperative manner by a previously unknown, adjacent second binding site. Mutations in the previously unknown site impaired PI(4,5)P2-dependent plasma membrane localization in living cells and PI(4,5)P2 interaction in silico, emphasizing its importance for PI(4,5)P2 affinity. The two-ligand binding mode may serve to sharpen the membrane association-dissociation cycle of tubby-like proteins that underlies delivery of ciliary cargo.
Collapse
Affiliation(s)
- Veronika Thallmair
- Institute for Physiology and Pathophysiology, Philipps University Marburg, Deutschhausstr. 1-2, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps University Marburg, Marburg, Germany
| | - Lea Schultz
- Institute for Physiology and Pathophysiology, Philipps University Marburg, Deutschhausstr. 1-2, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps University Marburg, Marburg, Germany
| | - Wencai Zhao
- Institute for Physiology and Pathophysiology, Philipps University Marburg, Deutschhausstr. 1-2, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps University Marburg, Marburg, Germany
| | - Siewert J. Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands
| | - Dominik Oliver
- Institute for Physiology and Pathophysiology, Philipps University Marburg, Deutschhausstr. 1-2, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps University Marburg, Marburg, Germany
- Corresponding author. (S.T.); (D.O.)
| | - Sebastian Thallmair
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, Netherlands
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Corresponding author. (S.T.); (D.O.)
| |
Collapse
|
13
|
Droubi A, Wallis C, Anderson KE, Rahman S, de Sa A, Rahman T, Stephens LR, Hawkins PT, Lowe M. The inositol 5-phosphatase INPP5B regulates B cell receptor clustering and signaling. J Cell Biol 2022; 221:e202112018. [PMID: 35878408 PMCID: PMC9351708 DOI: 10.1083/jcb.202112018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Upon antigen binding, the B cell receptor (BCR) undergoes clustering to form a signalosome that propagates downstream signaling required for normal B cell development and physiology. BCR clustering is dependent on remodeling of the cortical actin network, but the mechanisms that regulate actin remodeling in this context remain poorly defined. In this study, we identify the inositol 5-phosphatase INPP5B as a key regulator of actin remodeling, BCR clustering, and downstream signaling in antigen-stimulated B cells. INPP5B acts via dephosphorylation of the inositol lipid PI(4,5)P2 that in turn is necessary for actin disassembly, BCR mobilization, and cell spreading on immobilized surface antigen. These effects can be explained by increased actin severing by cofilin and loss of actin linking to the plasma membrane by ezrin, both of which are sensitive to INPP5B-dependent PI(4,5)P2 hydrolysis. INPP5B is therefore a new player in BCR signaling and may represent an attractive target for treatment of B cell malignancies caused by aberrant BCR signaling.
Collapse
Affiliation(s)
- Alaa Droubi
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Connor Wallis
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Saifur Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Aloka de Sa
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
14
|
Expression of the GFP-mammalian pleckstrin homology (PH) domain of the phospholipase C δ1 in Saccharomyces cerevisiae BY4741. Mol Biol Rep 2022; 49:4123-4128. [DOI: 10.1007/s11033-022-07414-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022]
|
15
|
Hammond GRV, Ricci MMC, Weckerly CC, Wills RC. An update on genetically encoded lipid biosensors. Mol Biol Cell 2022; 33:tp2. [PMID: 35420888 PMCID: PMC9282013 DOI: 10.1091/mbc.e21-07-0363] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 01/16/2023] Open
Abstract
Specific lipid species play central roles in cell biology. Their presence or enrichment in individual membranes can control properties or direct protein localization and/or activity. Therefore, probes to detect and observe these lipids in intact cells are essential tools in the cell biologist's freezer box. Herein, we discuss genetically encoded lipid biosensors, which can be expressed as fluorescent protein fusions to track lipids in living cells. We provide a state-of-the-art list of the most widely available and reliable biosensors and highlight new probes (circa 2018-2021). Notably, we focus on advances in biosensors for phosphatidylinositol, phosphatidic acid, and PI 3-kinase lipid products.
Collapse
Affiliation(s)
- Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Morgan M. C. Ricci
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Claire C. Weckerly
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Rachel C. Wills
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
16
|
Raut P, Weller SR, Obeng B, Soos BL, West BE, Potts CM, Sangroula S, Kinney MS, Burnell JE, King BL, Gosse JA, Hess ST. Cetylpyridinium chloride (CPC) reduces zebrafish mortality from influenza infection: Super-resolution microscopy reveals CPC interference with multiple protein interactions with phosphatidylinositol 4,5-bisphosphate in immune function. Toxicol Appl Pharmacol 2022; 440:115913. [PMID: 35149080 PMCID: PMC8824711 DOI: 10.1016/j.taap.2022.115913] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/04/2022] [Accepted: 02/04/2022] [Indexed: 01/12/2023]
Abstract
The COVID-19 pandemic raises significance for a potential influenza therapeutic compound, cetylpyridinium chloride (CPC), which has been extensively used in personal care products as a positively-charged quaternary ammonium antibacterial agent. CPC is currently in clinical trials to assess its effects on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) morbidity. Two published studies have provided mouse and human data indicating that CPC may alleviate influenza infection, and here we show that CPC (0.1 μM, 1 h) reduces zebrafish mortality and viral load following influenza infection. However, CPC mechanisms of action upon viral-host cell interaction are currently unknown. We have utilized super-resolution fluorescence photoactivation localization microscopy to probe the mode of CPC action. Reduction in density of influenza viral protein hemagglutinin (HA) clusters is known to reduce influenza infectivity: here, we show that CPC (at non-cytotoxic doses, 5-10 μM) reduces HA density and number of HA molecules per cluster within the plasma membrane of NIH-3T3 mouse fibroblasts. HA is known to colocalize with the negatively-charged mammalian lipid phosphatidylinositol 4,5-bisphosphate (PIP2); here, we show that nanoscale co-localization of HA with the PIP2-binding Pleckstrin homology (PH) reporter in the plasma membrane is diminished by CPC. CPC also dramatically displaces the PIP2-binding protein myristoylated alanine-rich C-kinase substrate (MARCKS) from the plasma membrane of rat RBL-2H3 mast cells; this disruption of PIP2 is correlated with inhibition of mast cell degranulation. Together, these findings offer a PIP2-focused mechanism underlying CPC disruption of influenza and suggest potential pharmacological use of this drug as an influenza therapeutic to reduce global deaths from viral disease.
Collapse
Affiliation(s)
- Prakash Raut
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Sasha R Weller
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Bright Obeng
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Brandy L Soos
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Bailey E West
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Christian M Potts
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Suraj Sangroula
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Marissa S Kinney
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - John E Burnell
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Benjamin L King
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Julie A Gosse
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA.
| | - Samuel T Hess
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA.
| |
Collapse
|
17
|
de la Cruz L, Kushmerick C, Sullivan JM, Kruse M, Vivas O. Hippocampal neurons maintain a large PtdIns(4)P pool that results in faster PtdIns(4,5)P2 synthesis. J Gen Physiol 2022; 154:e202113001. [PMID: 35179558 PMCID: PMC8906353 DOI: 10.1085/jgp.202113001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/01/2022] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
PtdIns(4,5)P2 is a signaling lipid central to the regulation of multiple cellular functions. It remains unknown how PtdIns(4,5)P2 fulfills various functions in different cell types, such as regulating neuronal excitability, synaptic release, and astrocytic function. Here, we compared the dynamics of PtdIns(4,5)P2 synthesis in hippocampal neurons and astrocytes with the kidney-derived tsA201 cell line. The experimental approach was to (1) measure the abundance and rate of PtdIns(4,5)P2 synthesis and precursors using specific biosensors, (2) measure the levels of PtdIns(4,5)P2 and its precursors using mass spectrometry, and (3) use a mathematical model to compare the metabolism of PtdIns(4,5)P2 in cell types with different proportions of phosphoinositides. The rate of PtdIns(4,5)P2 resynthesis in hippocampal neurons after depletion by cholinergic or glutamatergic stimulation was three times faster than for tsA201 cells. In tsA201 cells, resynthesis of PtdIns(4,5)P2 was dependent on the enzyme PI4K. In contrast, in hippocampal neurons, the resynthesis rate of PtdIns(4,5)P2 was insensitive to the inhibition of PI4K, indicating that it does not require de novo synthesis of the precursor PtdIns(4)P. Measurement of phosphoinositide abundance indicated a larger pool of PtdIns(4)P, suggesting that hippocampal neurons maintain sufficient precursor to restore PtdIns(4,5)P2 levels. Quantitative modeling indicates that the measured differences in PtdIns(4)P pool size and higher activity of PI4K can account for the experimental findings and indicates that high PI4K activity prevents depletion of PtdIns(4)P. We further show that the resynthesis of PtdIns(4,5)P2 is faster in neurons than astrocytes, providing context to the relevance of cell type-specific mechanisms to sustain PtdIns(4,5)P2 levels.
Collapse
Affiliation(s)
- Lizbeth de la Cruz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Christopher Kushmerick
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jane M. Sullivan
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Martin Kruse
- Department of Biology and Program in Neuroscience, Bates College, Lewiston, ME
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
18
|
Yerramilli VS, Ross AH, Scarlata S, Gericke A. IQGAP1 scaffolding links phosphoinositide kinases to cytoskeletal reorganization. Biophys J 2022; 121:793-807. [PMID: 35077666 PMCID: PMC8943696 DOI: 10.1016/j.bpj.2022.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/24/2021] [Accepted: 01/21/2022] [Indexed: 11/02/2022] Open
Abstract
IQGAP1 is a multidomain scaffold protein that coordinates the direction and impact of multiple signaling pathways by scaffolding its various binding partners. However, the spatial and temporal resolution of IQGAP1 scaffolding remains unclear. Here, we use fluorescence imaging and correlation methods that allow for real-time live-cell changes in IQGAP1 localization and complex formation during signaling. We find that IQGAP1 and PIPKIγ interact on both the plasma membrane and in cytosol. Epidermal growth factor (EGF) stimulation, which can initiate cytoskeletal changes, drives the movement of the cytosolic pool toward the plasma membrane to promote cytoskeletal changes. We also observe that a significant population of cytosolic IQGAP1-PIPKIγ complexes localize to early endosomes, and in some instances form aggregated clusters which become highly mobile upon EGF stimulation. Our imaging studies show that PIPKIγ and PI3K bind simultaneously to IQGAP1, which may accelerate conversion of PI4P to PI(3,4,5)P3 that is required for cytoskeletal changes. Additionally, we find that IQGAP1 is responsible for PIPKIγ association with two proteins associated with cytoskeletal changes, talin and Cdc42, during EGF stimulation. These results directly show that IQGAP1 provides a physical link between phosphoinositides (through PIPKIγ), focal adhesion formation (through talin), and cytoskeletal reorganization (through Cdc42) upon EGF stimulation. Taken together, our results support the importance of IQGAP1 in regulating cell migration by linking phosphoinositide lipid signaling with cytoskeletal reorganization.
Collapse
Affiliation(s)
- V. Siddartha Yerramilli
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Alonzo H. Ross
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Arne Gericke
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts.
| |
Collapse
|
19
|
Stilling S, Kalliakoudas T, Benninghoven-Frey H, Inoue T, Falkenburger BH. PIP2 determines length and stability of primary cilia by balancing membrane turnovers. Commun Biol 2022; 5:93. [PMID: 35079141 PMCID: PMC8789910 DOI: 10.1038/s42003-022-03028-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/23/2021] [Indexed: 11/09/2022] Open
Abstract
AbstractPrimary cilia are sensory organelles on many postmitotic cells. The ciliary membrane is continuous with the plasma membrane but differs in its phospholipid composition with phosphatidylinositol 4,5-bisposphate (PIP2) being much reduced toward the ciliary tip. In order to determine the functional significance of this difference, we used chemically induced protein dimerization to rapidly synthesize or degrade PIP2 selectively in the ciliary membrane. We observed ciliary fission when PIP2 was synthesized and a growing ciliary length when PIP2 was degraded. Ciliary fission required local actin polymerisation in the cilium, the Rho kinase Rac, aurora kinase A (AurkA) and histone deacetylase 6 (HDAC6). This pathway was previously described for ciliary disassembly before cell cycle re-entry. Activating ciliary receptors in the presence of dominant negative dynamin also increased ciliary PIP2, and the associated vesicle budding required ciliary PIP2. Finally, ciliary shortening resulting from constitutively increased ciliary PIP2 was mediated by the same actin – AurkA – HDAC6 pathway. Taken together, changes in ciliary PIP2 are a unifying point for ciliary membrane stability and turnover. Different stimuli increase ciliary PIP2 to secrete vesicles and reduce ciliary length by a common pathway. The paucity of PIP2 in the distal cilium therefore ensures ciliary stability.
Collapse
|
20
|
Ryan A, Hammond GRV, Deiters A. Optical Control of Phosphoinositide Binding: Rapid Activation of Subcellular Protein Translocation and Cell Signaling. ACS Synth Biol 2021; 10:2886-2895. [PMID: 34748306 DOI: 10.1021/acssynbio.1c00328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cells utilize protein translocation to specific compartments for spatial and temporal regulation of protein activity, in particular in the context of signaling processes. Protein recognition and binding to various subcellular membranes is mediated by a network of phosphatidylinositol phosphate (PIP) species bearing one or multiple phosphate moieties on the polar inositol head. Here, we report a new, highly efficient method for optical control of protein localization through the site-specific incorporation of a photocaged amino acid for steric and electrostatic disruption of inositol phosphate recognition and binding. We demonstrate general applicability of the approach by photocaging two unrelated proteins, sorting nexin 3 (SNX3) and the pleckstrin homology (PH) domain of phospholipase C delta 1 (PLCδ1), with two distinct PIP binding domains and distinct subcellular localizations. We have established the applicability of this methodology through its application to Son of Sevenless 2 (SOS2), a signaling protein involved in the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) cascade. Upon fusing the photocaged plasma membrane-targeted construct PH-enhanced green fluorescent protein (EGFP), to the catalytic domain of SOS2, we demonstrated light-induced membrane localization of the construct resulting in fast and extensive activation of the ERK signaling pathway in NIH 3T3 cells. This approach can be readily extended to other proteins, with minimal protein engineering, and provides a method for acute optical control of protein translocation with rapid and complete activation.
Collapse
Affiliation(s)
- Amy Ryan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
21
|
Sarmento MJ, Borges-Araújo L, Pinto SN, Bernardes N, Ricardo JC, Coutinho A, Prieto M, Fernandes F. Quantitative FRET Microscopy Reveals a Crucial Role of Cytoskeleton in Promoting PI(4,5)P 2 Confinement. Int J Mol Sci 2021; 22:11727. [PMID: 34769158 PMCID: PMC8583820 DOI: 10.3390/ijms222111727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/30/2023] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is an essential plasma membrane component involved in several cellular functions, including membrane trafficking and cytoskeleton organization. This function multiplicity is partially achieved through a dynamic spatiotemporal organization of PI(4,5)P2 within the membrane. Here, we use a Förster resonance energy transfer (FRET) approach to quantitatively assess the extent of PI(4,5)P2 confinement within the plasma membrane. This methodology relies on the rigorous evaluation of the dependence of absolute FRET efficiencies between pleckstrin homology domains (PHPLCδ) fused with fluorescent proteins and their average fluorescence intensity at the membrane. PI(4,5)P2 is found to be significantly compartmentalized at the plasma membrane of HeLa cells, and these clusters are not cholesterol-dependent, suggesting that membrane rafts are not involved in the formation of these nanodomains. On the other hand, upon inhibition of actin polymerization, compartmentalization of PI(4,5)P2 is almost entirely eliminated, showing that the cytoskeleton network is the critical component responsible for the formation of nanoscale PI(4,5)P2 domains in HeLa cells.
Collapse
Affiliation(s)
- Maria J. Sarmento
- Centro de Química-Física Molecular and Institute of Nanoscience and Nanotechnology, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal
| | - Luís Borges-Araújo
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Sandra N. Pinto
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Nuno Bernardes
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joana C. Ricardo
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
| | - Ana Coutinho
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Departamento de Química e Bioquímica, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Manuel Prieto
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Fábio Fernandes
- IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal; (L.B.-A.); (S.N.P.); (N.B.); (J.C.R.); (A.C.); (M.P.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal
| |
Collapse
|
22
|
de Souza LB, Ong HL, Liu X, Ambudkar IS. PIP 2 and septin control STIM1/Orai1 assembly by regulating cytoskeletal remodeling via a CDC42-WASP/WAVE-ARP2/3 protein complex. Cell Calcium 2021; 99:102475. [PMID: 34601312 DOI: 10.1016/j.ceca.2021.102475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/20/2022]
Abstract
Store-operated calcium entry (SOCE) is triggered by assembly of Orai1 with STIM proteins in ER-PM junctions. Plasma membrane PIP2 as well as PIP2-binding protein, SEPT4, significantly impact Orai1-STIM1 interaction. While septins and PIP2 can organize the actin cytoskeleton, it is unclear whether the status of actin within the junctions contributes to SOCE. We report herein that actin remodeling modulates STIM1 clustering. Our findings show that a PIP2- and SEPT4-dependent mechanism involving CDC42, WASP/WAVE, and ARP2 regulates actin remodeling into a ring-like structure around STIM1 puncta. CDC42 localization in the ER-plasma membrane region is enhanced following ER-Ca2+ store depletion. PIP2 depletion or knockdown of SEPT4 attenuate the recruitment of CDC42 to the ER-PM region. Importantly, knockdown of SEPT4, or CDC42+ARP2, disrupts the organization of actin as well as STIM1 clustering. Consequently, Orai1 recruitment to STIM1 puncta, SOCE, and NFAT translocation to the nucleus are all attenuated. Ca2+ influx induced by STIM1-C terminus is not affected by CDC42 knockdown. In aggregate, our findings reveal that PIP2 and SEPT4 affect Orai1/STIM1 clustering by coordinating actin remodeling within ER-PM junctions. This dynamic reorganization of actin has an important role in regulation of SOCE and downstream Ca2+-dependent effector functions.
Collapse
Affiliation(s)
- Lorena Brito de Souza
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA
| | - Hwei Ling Ong
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA.
| | - Xibao Liu
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA
| | - Indu S Ambudkar
- Secretory Physiology Section, NIDCR, NIH, Bldg. 10/Room 1N-113, Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Myeong J, de la Cruz L, Jung SR, Yeon JH, Suh BC, Koh DS, Hille B. Phosphatidylinositol 4,5-bisphosphate is regenerated by speeding of the PI 4-kinase pathway during long PLC activation. J Gen Physiol 2021; 152:211533. [PMID: 33186442 PMCID: PMC7671494 DOI: 10.1085/jgp.202012627] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/13/2020] [Indexed: 01/05/2023] Open
Abstract
The dynamic metabolism of membrane phosphoinositide lipids involves several cellular compartments including the ER, Golgi, and plasma membrane. There are cycles of phosphorylation and dephosphorylation and of synthesis, transfer, and breakdown. The simplified phosphoinositide cycle comprises synthesis of phosphatidylinositol in the ER, transport, and phosphorylation in the Golgi and plasma membranes to generate phosphatidylinositol 4,5-bisphosphate, followed by receptor-stimulated hydrolysis in the plasma membrane and return of the components to the ER for reassembly. Using probes for specific lipid species, we have followed and analyzed the kinetics of several of these events during stimulation of M1 muscarinic receptors coupled to the G-protein Gq. We show that during long continued agonist action, polyphosphorylated inositol lipids are initially depleted but then regenerate while agonist is still present. Experiments and kinetic modeling reveal that the regeneration results from gradual but massive up-regulation of PI 4-kinase pathways rather than from desensitization of receptors. Golgi pools of phosphatidylinositol 4-phosphate and the lipid kinase PI4KIIIα (PI4KA) contribute to this homeostatic regeneration. This powerful acceleration, which may be at the level of enzyme activity or of precursor and product delivery, reveals strong regulatory controls in the phosphoinositide cycle.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | - Lizbeth de la Cruz
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | | | - Jun-Hee Yeon
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Byung-Chang Suh
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Duk-Su Koh
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | - Bertil Hille
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
24
|
Sharma P, Yadav SK, Shah SD, Javed E, Lim JM, Pan S, Nayak AP, Panettieri RA, Penn RB, Kambayashi T, Deshpande DA. Diacylglycerol Kinase Inhibition Reduces Airway Contraction by Negative Feedback Regulation of Gq-signaling. Am J Respir Cell Mol Biol 2021; 65:658-671. [PMID: 34293268 DOI: 10.1165/rcmb.2021-0106oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Exaggerated airway smooth muscle (ASM) contraction regulated by the Gq family of G protein-coupled receptors (GPCRs) causes airway hyperresponsiveness (AHR) in asthma. Activation of Gq-coupled GPCRs leads to phospholipase C (PLC)-mediated generation of inositol triphosphate (IP3) and diacylglycerol (DAG). DAG signaling is terminated by the action of DAG kinase (DGK) that converts DAG into phosphatidic acid (PA). Our previous study demonstrated that DGKα and ζ isoform knockout mice are protected from the development of allergen-induced AHR. Here we aimed at determining the mechanism by which DGK regulates ASM contraction. Activity of DGK isoforms was inhibited in human ASM cells by siRNA-mediated knockdown of DGKα and ζwhile pharmacological inhibition was achieved by pan DGK inhibitor I (R59022). Effects of DGK inhibition on contractile agonist-induced activation of PLC and myosin light chain (MLC) kinase, elevation of IP3, and calcium levels were assessed. Further, we employed human precision-cut lung slices and assessed the role of DGK in agonist-induced bronchoconstriction. DGK inhibitor I attenuated histamine- and methacholine-induced bronchoconstriction. DGKα and ζ knockdown or pre-treatment with DGK inhibitor I resulted in attenuated agonist-induced phosphorylation of MLC and myosin light chain phosphatase in ASM cells. Further, DGK inhibition decreased Gq agonist-induced calcium elevation, generation of IP3, and increased histamine-induced production of PA. Finally, DGK inhibition or treatment with DAG analog resulted in attenuation of activation of PLC in human ASM cells. Our findings suggest that DGK inhibition perturbed the DAG:PA ratio resulting in inhibition of Gq-PLC activation in a negative feedback manner, resulting in protection against ASM contraction.
Collapse
Affiliation(s)
- Pawan Sharma
- Thomas Jefferson University - Center City Campus, 6559, Medicine, Philadelphia, Pennsylvania, United States.,University of Tasmania Faculty of Health, 60119, Hobart, Tasmania, Australia
| | - Santosh K Yadav
- Thomas Jefferson University, 6559, Medicine, Philadelphia, Pennsylvania, United States
| | - Sushrut D Shah
- Thomas Jefferson University, 6559, Medicine, Philadelphia, Pennsylvania, United States
| | - Elham Javed
- Thomas Jefferson University, 6559, Medicine, Philadelphia, Pennsylvania, United States
| | - John M Lim
- Thomas Jefferson University Sidney Kimmel Medical College, 12313, Philadelphia, Pennsylvania, United States
| | - Shi Pan
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Ajay P Nayak
- Thomas Jefferson University, 6559, Medicine, Philadelphia, Pennsylvania, United States
| | - Reynold A Panettieri
- Rutgers University, 242612, Rutgers Institute for Translational Medicine and Science, Child Health Institute, New Brunswick, New Jersey, United States
| | - Raymond B Penn
- Thomas Jefferson University, 6559, Medicine, Philadelphia, Pennsylvania, United States
| | - Taku Kambayashi
- University of Pennsylvania, 6572, Pathology, Philadelphia, Pennsylvania, United States
| | - Deepak A Deshpande
- Thomas Jefferson University, 6559, Center for Translational Medicine, Philadelphia, Pennsylvania, United States;
| |
Collapse
|
25
|
de Jong F, Munnik T. Attracted to membranes: lipid-binding domains in plants. PLANT PHYSIOLOGY 2021; 185:707-723. [PMID: 33793907 PMCID: PMC8133573 DOI: 10.1093/plphys/kiaa100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/11/2020] [Indexed: 05/18/2023]
Abstract
Membranes are essential for cells and organelles to function. As membranes are impermeable to most polar and charged molecules, they provide electrochemical energy to transport molecules across and create compartmentalized microenvironments for specific enzymatic and cellular processes. Membranes are also responsible for guided transport of cargoes between organelles and during endo- and exocytosis. In addition, membranes play key roles in cell signaling by hosting receptors and signal transducers and as substrates and products of lipid second messengers. Anionic lipids and their specific interaction with target proteins play an essential role in these processes, which are facilitated by specific lipid-binding domains. Protein crystallography, lipid-binding studies, subcellular localization analyses, and computer modeling have greatly advanced our knowledge over the years of how these domains achieve precision binding and what their function is in signaling and membrane trafficking, as well as in plant development and stress acclimation.
Collapse
Affiliation(s)
- Femke de Jong
- Cluster Green Life Sciences, Section Plant Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Teun Munnik
- Cluster Green Life Sciences, Section Plant Cell Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Abstract
Ion channel are embedded in the lipid bilayers of biological membranes. Membrane phospholipids constitute a barrier to ion movement, and they have been considered for a long time as a passive environment for channel proteins. Membrane phospholipids, however, do not only serve as a passive amphipathic environment, but they also modulate channel activity by direct specific lipid-protein interactions. Phosphoinositides are quantitatively minor components of biological membranes, and they play roles in many cellular functions, including membrane traffic, cellular signaling and cytoskeletal organization. Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is mainly found in the inner leaflet of the plasma membrane. Its role as a potential ion channel regulator was first appreciated over two decades ago and by now this lipid is a well-established cofactor or regulator of many different ion channels. The past two decades witnessed the steady development of techniques to study ion channel regulation by phosphoinositides with progress culminating in recent cryoEM structures that allowed visualization of how PI(4,5)P2 opens some ion channels. This chapter will provide an overview of the methods to study regulation by phosphoinositides, focusing on plasma membrane ion channels and PI(4,5)P2.
Collapse
|
27
|
Ashraf APK, Gerke V. Plasma membrane wound repair is characterized by extensive membrane lipid and protein rearrangements in vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118991. [PMID: 33667528 DOI: 10.1016/j.bbamcr.2021.118991] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 11/28/2022]
Abstract
Vascular endothelial cells are subject to mechanical stress resulting from blood flow and interactions with leukocytes. Stress occurs at the apical, vessel-facing cell surface and leads to membrane ruptures that have to be resealed to ensure cell survival. To mimic this process, we developed a laser ablation protocol selectively inducing wounds in the apical plasma membrane of endothelial cells. We show that Ca2+-dependent membrane resealing is initiated following this wounding protocol and that the process is accompanied by substantial membrane lipid dynamics at the wound site. Specifically, phosphatidylinositol (4,5)-bisphosphate, phosphatidylserine and phosphatidic acid rapidly accumulate at membrane wounds forming potential interaction platforms for Ca2+/phospholipid binding proteins of the annexin (Anx) family that are also recruited within seconds after wounding. Depletion of one annexin, AnxA2, and its putative binding partner S100A11 interferes with membrane resealing suggesting that Ca2+-dependent annexin-phospholipid interactions are required for efficient membrane wound repair in endothelial cells.
Collapse
Affiliation(s)
- Arsila P K Ashraf
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Strasse 56, 48149, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Strasse 56, 48149, Münster, Germany.
| |
Collapse
|
28
|
Abstract
Lipids, like phosphoinositides, can be visualized in living cells in real time using genetically encoded biosensors and fluorescence microscopy. Sensor localization can be quantified by determining the fluorescence intensity of each fluorophore. Enrichment of lipids at membranes can be determined by generating and applying an organelle-specific binary mask. In this chapter, we provide a detailed list of reagents and methods to visualize and quantify relative lipid levels. Applying this approach, changes in lipid levels can be assessed in cases when lipid metabolizing enzymes are mutated or otherwise altered.
Collapse
Affiliation(s)
- Rachel C Wills
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Pacheco
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Wen Y, Feigenson GW, Vogt VM, Dick RA. Mechanisms of PI(4,5)P2 Enrichment in HIV-1 Viral Membranes. J Mol Biol 2020; 432:5343-5364. [PMID: 32739462 PMCID: PMC8262684 DOI: 10.1016/j.jmb.2020.07.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/12/2020] [Accepted: 07/26/2020] [Indexed: 01/10/2023]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) is critical for HIV-1 virus assembly. The viral membrane is enriched in PIP2, suggesting that the virus assembles at PIP2-rich microdomains. We showed previously that in model membranes PIP2 can form nanoscopic clusters bridged by multivalent cations. Here, using purified proteins we quantitated the binding of HIV-1 Gag-related proteins to giant unilamellar vesicles containing either clustered or free PIP2. Myristoylated MA strongly preferred binding to clustered PIP2. By contrast, unmyristoylated HIV-1 MA, RSV MA, and a PH domain all preferred to interact with free PIP2. We also found that HIV-1 Gag multimerization promotes PIP2 clustering. Truncated Gag proteins comprising the MA, CA, and SP domains (MACASP) or the MA and CA domains (MACA) induced self-quenching of acyl chain-labeled fluorescent PIP2 in liposomes, implying clustering. However, HIV-1 MA itself did not induce PIP2 clustering. A CA inter-hexamer dimer interface mutation led to a loss of induced PIP2 clustering in MACA, indicating the importance of protein multimerization. Cryo-electron tomography of liposomes with bound MACA showed an amorphous protein layer on the membrane surface. Thus, it appears that while protein–protein interactions are required for PIP2 clustering, formation of a regular lattice is not. Protein-induced PIP2 clustering and multivalent cation-induced PIP2 clustering are additive. Taken together, these results provide the first evidence that HIV-1 Gag can selectively target pre-existing PIP2-enriched domains of the plasma membrane for viral assembly, and that Gag multimerization can further enrich PIP2 at assembly sites. These effects could explain the observed PIP2 enrichment in HIV-1.
Collapse
Affiliation(s)
- Yi Wen
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Gerald W Feigenson
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Volker M Vogt
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Robert A Dick
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
30
|
STIM2 targets Orai1/STIM1 to the AKAP79 signaling complex and confers coupling of Ca 2+ entry with NFAT1 activation. Proc Natl Acad Sci U S A 2020; 117:16638-16648. [PMID: 32601188 DOI: 10.1073/pnas.1915386117] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Orai1 channel is regulated by stromal interaction molecules STIM1 and STIM2 within endoplasmic reticulum (ER)-plasma membrane (PM) contact sites. Ca2+ signals generated by Orai1 activate Ca2+-dependent gene expression. When compared with STIM1, STIM2 is a weak activator of Orai1, but it has been suggested to have a unique role in nuclear factor of activated T cells 1 (NFAT1) activation triggered by Orai1-mediated Ca2+ entry. In this study, we examined the contribution of STIM2 in NFAT1 activation. We report that STIM2 recruitment of Orai1/STIM1 to ER-PM junctions in response to depletion of ER-Ca2+ promotes assembly of the channel with AKAP79 to form a signaling complex that couples Orai1 channel function to the activation of NFAT1. Knockdown of STIM2 expression had relatively little effect on Orai1/STIM1 clustering or local and global [Ca2+]i increases but significantly attenuated NFAT1 activation and assembly of Orai1 with AKAP79. STIM1ΔK, which lacks the PIP2-binding polybasic domain, was recruited to ER-PM junctions following ER-Ca2+ depletion by binding to Orai1 and caused local and global [Ca2+]i increases comparable to those induced by STIM1 activation of Orai1. However, in contrast to STIM1, STIM1ΔK induced less NFAT1 activation and attenuated the association of Orai1 with STIM2 and AKAP79. Orai1-AKAP79 interaction and NFAT1 activation were recovered by coexpressing STIM2 with STIM1ΔK. Replacing the PIP2-binding domain of STIM1 with that of STIM2 eliminated the requirement of STIM2 for NFAT1 activation. Together, these data demonstrate an important role for STIM2 in coupling Orai1-mediated Ca2+ influx to NFAT1 activation.
Collapse
|
31
|
Finkelstein S, Gospe SM, Schuhmann K, Shevchenko A, Arshavsky VY, Lobanova ES. Phosphoinositide Profile of the Mouse Retina. Cells 2020; 9:cells9061417. [PMID: 32517352 PMCID: PMC7349851 DOI: 10.3390/cells9061417] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
Phosphoinositides are known to play multiple roles in eukaryotic cells. Although dysregulation of phosphoinositide metabolism in the retina has been reported to cause visual dysfunction in animal models and human patients, our understanding of the phosphoinositide composition of the retina is limited. Here, we report a characterization of the phosphoinositide profile of the mouse retina and an analysis of the subcellular localization of major phosphorylated phosphoinositide forms in light-sensitive photoreceptor neurons. Using chromatography of deacylated phosphatidylinositol headgroups, we established PI(4,5)P2 and PI(4)P as two major phosphorylated phosphoinositides in the retina. Using high-resolution mass spectrometry, we revealed 18:0/20:4 and 16:0/20:4 as major fatty-acyl chains of retinal phosphoinositides. Finally, analysis of fluorescent phosphoinositide sensors in rod photoreceptors demonstrated distinct subcellular distribution patterns of major phosphoinositides. The PI(4,5)P2 reporter was enriched in the inner segments and synapses, but was barely detected in the light-sensitive outer segments. The PI(4)P reporter was mostly found in the outer and inner segments and the areas around nuclei, but to a lesser degree in the synaptic region. These findings provide support for future mechanistic studies defining the biological significance of major mono- (PI(4)P) and bisphosphate (PI(4,5)P2) phosphatidylinositols in photoreceptor biology and retinal health.
Collapse
Affiliation(s)
- Stella Finkelstein
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
| | - Sidney M. Gospe
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
| | - Kai Schuhmann
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; (K.S.); (A.S.)
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; (K.S.); (A.S.)
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Ekaterina S. Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
- Correspondence:
| |
Collapse
|
32
|
Archer CR, Enslow BT, Carver CM, Stockand JD. Phosphatidylinositol 4,5-bisphosphate directly interacts with the β and γ subunits of the sodium channel ENaC. J Biol Chem 2020; 295:7958-7969. [PMID: 32341072 PMCID: PMC7278353 DOI: 10.1074/jbc.ra120.012606] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
The plasma membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) regulates the activity of diverse ion channels to include the epithelial Na+ channel ENaC. Whether PIP2 regulation of ENaC is due to a direct phospholipid-protein interaction, remains obscure. To date, possible interaction of PIP2 with ENaC primarily has been tested indirectly through assays of channel function. A fragment-based biochemical analysis approach is used here to directly quantify possible PIP2-ENaC interactions. We find using the CIBN-CRY2 optogenetic dimerization system that the phosphoryl group positioned at carbon 5 of PIP2 is necessary for interaction with ENaC. Previous studies have implicated conserved basic residues in the cytosolic portions of β- and γ-ENaC subunits as being important for PIP2-ENaC interactions. To test this, we used synthetic peptides of these regions of β- and γ-ENaC. Steady-state intrinsic fluorescence spectroscopy demonstrated that phosphoinositides change the local conformation of the N terminus of β-ENaC, and two sites of γ-ENaC adjacent to the plasma membrane, suggesting direct interactions of PIP2 with these three regions. Microscale thermophoresis elaborated PIP2 interactions with the N termini of β- (Kd ∼5.2 μm) and γ-ENaC (Kd ∼13 μm). A weaker interaction site within the carboxyl terminus of γ-ENaC (Kd ∼800 μm) was also observed. These results support that PIP2 regulates ENaC activity by directly interacting with at least three distinct regions within the cytoplasmic domains of the channel that contain conserved basic residues. These interactions are probably electrostatic in nature, and are likely to bear a key structural role in support of channel activity.
Collapse
Affiliation(s)
- Crystal R Archer
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, United States
| | - Benjamin T Enslow
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, United States
| | - Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, United States
| | - James D Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, United States
| |
Collapse
|
33
|
Martín-Aragón Baudel MAS, Shi J, Large WA, Albert AP. Insights into Activation Mechanisms of Store-Operated TRPC1 Channels in Vascular Smooth Muscle. Cells 2020; 9:E179. [PMID: 31936855 PMCID: PMC7017204 DOI: 10.3390/cells9010179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 01/10/2023] Open
Abstract
In vascular smooth muscle cells (VMSCs), the stimulation of store-operated channels (SOCs) mediate Ca2+ influx pathways which regulate important cellular functions including contraction, proliferation, migration, and growth that are associated with the development of vascular diseases. It is therefore important that we understand the biophysical, molecular composition, activation pathways, and physiological significance of SOCs in VSMCs as these maybe future therapeutic targets for conditions such as hypertension and atherosclerosis. Archetypal SOCs called calcium release-activated channels (CRACs) are composed of Orai1 proteins and are stimulated by the endo/sarcoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1) following store depletion. In contrast, this review focuses on proposals that canonical transient receptor potential (TRPC) channels composed of a heteromeric TRPC1/C5 molecular template, with TRPC1 conferring activation by store depletion, mediate SOCs in native contractile VSMCs. In particular, it summarizes our recent findings which describe a novel activation pathway of these TRPC1-based SOCs, in which protein kinase C (PKC)-dependent TRPC1 phosphorylation and phosphatidylinositol 4,5-bisphosphate (PIP2) are obligatory for channel opening. This PKC- and PIP2-mediated gating mechanism is regulated by the PIP2-binding protein myristoylated alanine-rich C kinase (MARCKS) and is coupled to store depletion by TRPC1-STIM1 interactions which induce Gq/PLCβ1 activity. Interestingly, the biophysical properties and activation mechanisms of TRPC1-based SOCs in native contractile VSMCs are unlikely to involve Orai1.
Collapse
Affiliation(s)
| | - Jian Shi
- LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK;
| | - William A. Large
- Vascular Biology Research Centre, Molecular and Clinical Research Institute, St. George’s, University of London, London SW17 0RE, UK;
| | - Anthony P. Albert
- Vascular Biology Research Centre, Molecular and Clinical Research Institute, St. George’s, University of London, London SW17 0RE, UK;
| |
Collapse
|
34
|
Nocita E, Del Giovane A, Tiberi M, Boccuni L, Fiorelli D, Sposato C, Romano E, Basoli F, Trombetta M, Rainer A, Traversa E, Ragnini-Wilson A. EGFR/ErbB Inhibition Promotes OPC Maturation up to Axon Engagement by Co-Regulating PIP2 and MBP. Cells 2019; 8:cells8080844. [PMID: 31390799 PMCID: PMC6721729 DOI: 10.3390/cells8080844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022] Open
Abstract
Remyelination in the adult brain relies on the reactivation of the Neuronal Precursor Cell (NPC) niche and differentiation into Oligodendrocyte Precursor Cells (OPCs) as well as on OPC maturation into myelinating oligodendrocytes (OLs). These two distinct phases in OL development are defined by transcriptional and morphological changes. How this differentiation program is controlled remains unclear. We used two drugs that stimulate myelin basic protein (MBP) expression (Clobetasol and Gefitinib) alone or combined with epidermal growth factor receptor (EGFR) or Retinoid X Receptor gamma (RXRγ) gene silencing to decode the receptor signaling required for OPC differentiation in myelinating OLs. Electrospun polystyrene (PS) microfibers were used as synthetic axons to study drug efficacy on fiber engagement. We show that EGFR inhibition per se stimulates MBP expression and increases Clobetasol efficacy in OPC differentiation. Consistent with this, Clobetasol and Gefitinib co-treatment, by co-regulating RXRγ, MBP and phosphatidylinositol 4,5-bisphosphate (PIP2) levels, maximizes synthetic axon engagement. Conversely, RXRγ gene silencing reduces the ability of the drugs to promote MBP expression. This work provides a view of how EGFR/ErbB inhibition controls OPC differentiation and indicates the combination of Clobetasol and Gefitinib as a potent remyelination-enhancing treatment.
Collapse
Affiliation(s)
- Emanuela Nocita
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Alice Del Giovane
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Marta Tiberi
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Laura Boccuni
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Denise Fiorelli
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Carola Sposato
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Elena Romano
- Advanced Microscopy Center, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Francesco Basoli
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Marcella Trombetta
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Enrico Traversa
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Antonella Ragnini-Wilson
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
35
|
Mondin VE, Ben El Kadhi K, Cauvin C, Jackson-Crawford A, Bélanger E, Decelle B, Salomon R, Lowe M, Echard A, Carréno S. PTEN reduces endosomal PtdIns(4,5)P 2 in a phosphatase-independent manner via a PLC pathway. J Cell Biol 2019; 218:2198-2214. [PMID: 31118240 PMCID: PMC6605811 DOI: 10.1083/jcb.201805155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 03/15/2019] [Accepted: 05/02/2019] [Indexed: 01/14/2023] Open
Abstract
This work reveals that the tumor suppressor PTEN acts through a PLC to reduce levels of endosomal PtdIns(4,5)P2, its own enzymatic product. This pathway can be chemically activated to rescue OCRL1 depletion in several disease models of the Lowe syndrome, a rare multisystemic genetic disease. The tumor suppressor PTEN dephosphorylates PtdIns(3,4,5)P3 into PtdIns(4,5)P2. Here, we make the unexpected discovery that in Drosophila melanogaster PTEN reduces PtdIns(4,5)P2 levels on endosomes, independently of its phosphatase activity. This new PTEN function requires the enzymatic action of dPLCXD, an atypical phospholipase C. Importantly, we discovered that this novel PTEN/dPLCXD pathway can compensate for depletion of dOCRL, a PtdIns(4,5)P2 phosphatase. Mutation of OCRL1, the human orthologue of dOCRL, causes oculocerebrorenal Lowe syndrome, a rare multisystemic genetic disease. Both OCRL1 and dOCRL loss have been shown to promote accumulation of PtdIns(4,5)P2 on endosomes and cytokinesis defects. Here, we show that PTEN or dPLCXD overexpression prevents these defects. In addition, we found that chemical activation of this pathway restores normal cytokinesis in human Lowe syndrome cells and rescues OCRL phenotypes in a zebrafish Lowe syndrome model. Our findings identify a novel PTEN/dPLCXD pathway that controls PtdIns(4,5)P2 levels on endosomes. They also point to a potential new strategy for the treatment of Lowe syndrome.
Collapse
Affiliation(s)
- Virginie E Mondin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Khaled Ben El Kadhi
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Clothilde Cauvin
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, Centre National de la Recherche Scientifique, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | | | - Emilie Bélanger
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Barbara Decelle
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Rémi Salomon
- Institut des Maladies Génétiques Imagine, Hôpital Necker-Enfants Malades, Université Paris Descartes, Paris, France
| | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, Centre National de la Recherche Scientifique, Paris, France
| | - Sébastien Carréno
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada .,Université de Montréal, Département de Pathologie et de Biologie Cellulaire, Montreal, Canada
| |
Collapse
|
36
|
Adriaans IE, Basant A, Ponsioen B, Glotzer M, Lens SM. PLK1 plays dual roles in centralspindlin regulation during cytokinesis. J Cell Biol 2019; 218:1250-1264. [PMID: 30728176 PMCID: PMC6446842 DOI: 10.1083/jcb.201805036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/26/2018] [Accepted: 01/23/2019] [Indexed: 11/26/2022] Open
Abstract
Cytokinesis begins upon anaphase onset. An early step involves local activation of the small GTPase RhoA, which triggers assembly of an actomyosin-based contractile ring at the equatorial cortex. Here, we delineated the contributions of PLK1 and Aurora B to RhoA activation and cytokinesis initiation in human cells. Knock-down of PRC1, which disrupts the spindle midzone, revealed the existence of two pathways that can initiate cleavage furrow ingression. One pathway depends on a well-organized spindle midzone and PLK1, while the other depends on Aurora B activity and centralspindlin at the equatorial cortex and can operate independently of PLK1. We further show that PLK1 inhibition sequesters centralspindlin onto the spindle midzone, making it unavailable for Aurora B at the equatorial cortex. We propose that PLK1 activity promotes the release of centralspindlin from the spindle midzone through inhibition of PRC1, allowing centralspindlin to function as a regulator of spindle midzone formation and as an activator of RhoA at the equatorial cortex.
Collapse
Affiliation(s)
- Ingrid E. Adriaans
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Angika Basant
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL
| | - Bas Ponsioen
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL
| | - Susanne M.A. Lens
- Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
Abstract
Lipids convey both structural and functional properties to eukaryotic membranes. Understanding the basic lipid composition and the dynamics of these important molecules, in the context of cellular membranes, can shed light on signaling, metabolism, trafficking, and even membrane identity. The development of genetically encoded lipid biosensors has allowed for the visualization of specific lipids inside individual, living cells. However, a number of caveats and considerations have emerged with the overexpression of these biosensors. In this Technical Perspective, we provide a current list of available genetically encoded lipid biosensors, together with criteria that determine their veracity. We also provide some suggestions for the optimal utilization of these biosensors when both designing experiments and interpreting results.
Collapse
Affiliation(s)
- Rachel C Wills
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 16261
| | - Brady D Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 16261
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 16261
| |
Collapse
|
38
|
Glantz ST, Berlew EE, Chow BY. Synthetic cell-like membrane interfaces for probing dynamic protein-lipid interactions. Methods Enzymol 2019; 622:249-270. [PMID: 31155055 DOI: 10.1016/bs.mie.2019.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The ability to rapidly screen interactions between proteins and membrane-like interfaces would aid in establishing the structure-function of protein-lipid interactions, provide a platform for engineering lipid-interacting protein tools, and potentially inform the signaling mechanisms and dynamics of membrane-associated proteins. Here, we describe the preparation and application of water-in-oil (w/o) emulsions with lipid-stabilized droplet interfaces that emulate the plasma membrane inner leaflet with tunable composition. Fluorescently labeled proteins are easily visualized in these synthetic cell-like droplets on an automated inverted fluorescence microscope, thus allowing for both rapid screening of relative binding and spatiotemporally resolved analyses of for example, protein-interface association and dissociation dynamics and competitive interactions, using commonplace instrumentation. We provide protocols for droplet formation, automated imaging assays and analysis, and the production of the positive control protein BcLOV4, a natural photoreceptor with a directly light-regulated interaction with anionic membrane phospholipids that is useful for optogenetic membrane recruitment.
Collapse
Affiliation(s)
- Spencer T Glantz
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Erin E Berlew
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Brian Y Chow
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
39
|
IPIP27 Coordinates PtdIns(4,5)P 2 Homeostasis for Successful Cytokinesis. Curr Biol 2019; 29:775-789.e7. [PMID: 30799246 PMCID: PMC6408333 DOI: 10.1016/j.cub.2019.01.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/03/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
During cytokinesis, an actomyosin contractile ring drives the separation of the two daughter cells. A key molecule in this process is the inositol lipid PtdIns(4,5)P2, which recruits numerous factors to the equatorial region for contractile ring assembly. Despite the importance of PtdIns(4,5)P2 in cytokinesis, the regulation of this lipid in cell division remains poorly understood. Here, we identify a role for IPIP27 in mediating cellular PtdIns(4,5)P2 homeostasis. IPIP27 scaffolds the inositol phosphatase oculocerebrorenal syndrome of Lowe (OCRL) by coupling it to endocytic BAR domain proteins. Loss of IPIP27 causes accumulation of PtdIns(4,5)P2 on aberrant endomembrane vacuoles, mislocalization of the cytokinetic machinery, and extensive cortical membrane blebbing. This phenotype is observed in Drosophila and human cells and can result in cytokinesis failure. We have therefore identified IPIP27 as a key modulator of cellular PtdIns(4,5)P2 homeostasis required for normal cytokinesis. The results indicate that scaffolding of inositol phosphatase activity is critical for maintaining PtdIns(4,5)P2 homeostasis and highlight a critical role for this process in cell division. IPIP27 scaffolds the inositol phosphatase OCRL via coupling to BAR domain proteins IPIP27 scaffolding of OCRL is critical for cellular PtdIns(4,5)P2 homeostasis IPIP27 is required for cortical actin and membrane stability during cytokinesis IPIP27 function is conserved from flies to humans
Collapse
|
40
|
Pemberton JG, Balla T. Polyphosphoinositide-Binding Domains: Insights from Peripheral Membrane and Lipid-Transfer Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1111:77-137. [PMID: 30483964 DOI: 10.1007/5584_2018_288] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Within eukaryotic cells, biochemical reactions need to be organized on the surface of membrane compartments that use distinct lipid constituents to dynamically modulate the functions of integral proteins or influence the selective recruitment of peripheral membrane effectors. As a result of these complex interactions, a variety of human pathologies can be traced back to improper communication between proteins and membrane surfaces; either due to mutations that directly alter protein structure or as a result of changes in membrane lipid composition. Among the known structural lipids found in cellular membranes, phosphatidylinositol (PtdIns) is unique in that it also serves as the membrane-anchored precursor of low-abundance regulatory lipids, the polyphosphoinositides (PPIn), which have restricted distributions within specific subcellular compartments. The ability of PPIn lipids to function as signaling platforms relies on both non-specific electrostatic interactions and the selective stereospecific recognition of PPIn headgroups by specialized protein folds. In this chapter, we will attempt to summarize the structural diversity of modular PPIn-interacting domains that facilitate the reversible recruitment and conformational regulation of peripheral membrane proteins. Outside of protein folds capable of capturing PPIn headgroups at the membrane interface, recent studies detailing the selective binding and bilayer extraction of PPIn species by unique functional domains within specific families of lipid-transfer proteins will also be highlighted. Overall, this overview will help to outline the fundamental physiochemical mechanisms that facilitate localized interactions between PPIn lipids and the wide-variety of PPIn-binding proteins that are essential for the coordinate regulation of cellular metabolism and membrane dynamics.
Collapse
Affiliation(s)
- Joshua G Pemberton
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
41
|
de Rubio RG, Ransom RF, Malik S, Yule DI, Anantharam A, Smrcka AV. Phosphatidylinositol 4-phosphate is a major source of GPCR-stimulated phosphoinositide production. Sci Signal 2018; 11:11/547/eaan1210. [PMID: 30206135 DOI: 10.1126/scisignal.aan1210] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Phospholipase C (PLC) enzymes hydrolyze the plasma membrane (PM) lipid phosphatidylinositol 4,5-bisphosphate (PI4,5P2) to generate the second messengers inositol trisphosphate (IP3) and diacylglycerol (DAG) in response to receptor activation in almost all mammalian cells. We previously found that stimulation of G protein-coupled receptors (GPCRs) in cardiac cells leads to the PLC-dependent hydrolysis of phosphatidylinositol 4-phosphate (PI4P) at the Golgi, a process required for the activation of nuclear protein kinase D (PKD) during cardiac hypertrophy. We hypothesized that GPCR-stimulated PLC activation leading to direct PI4P hydrolysis may be a general mechanism for DAG production. We measured GPCR activation-dependent changes in PM and Golgi PI4P pools in various cells using GFP-based detection of PI4P. Stimulation with various agonists caused a time-dependent reduction in PI4P-associated, but not PI4,5P2-associated, fluorescence at the Golgi and PM. Targeted depletion of PI4,5P2 from the PM before GPCR stimulation had no effect on the depletion of PM or Golgi PI4P, total inositol phosphate (IP) production, or PKD activation. In contrast, acute depletion of PI4P specifically at the PM completely blocked the GPCR-dependent production of IPs and activation of PKD but did not change the abundance of PI4,5P2 Acute depletion of Golgi PI4P had no effect on these processes. These data suggest that most of the PM PI4,5P2 pool is not involved in GPCR-stimulated phosphoinositide hydrolysis and that PI4P at the PM is responsible for the bulk of receptor-stimulated phosphoinositide hydrolysis and DAG production.
Collapse
Affiliation(s)
- Rafael Gil de Rubio
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Richard F Ransom
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Arun Anantharam
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA. .,Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
42
|
Gawden-Bone CM, Frazer GL, Richard AC, Ma CY, Strege K, Griffiths GM. PIP5 Kinases Regulate Membrane Phosphoinositide and Actin Composition for Targeted Granule Secretion by Cytotoxic Lymphocytes. Immunity 2018; 49:427-437.e4. [PMID: 30217409 PMCID: PMC6162341 DOI: 10.1016/j.immuni.2018.08.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 06/22/2018] [Accepted: 08/21/2018] [Indexed: 01/19/2023]
Abstract
How cytotoxic T lymphocytes (CTLs) sense T cell receptor (TCR) signaling in order to specialize an area of plasma membrane for granule secretion is not understood. Here, we demonstrate that immune synapse formation led to rapid localized changes in the phosphoinositide composition of the plasma membrane, both reducing phosphoinositide-4-phosphate (PI(4)P), PI(4,5)P2, and PI(3,4,5)P3 and increasing diacylglycerol (DAG) and PI(3,4)P2 within the first 2 min of synapse formation. These changes reduced negative charge across the synapse, triggering the release of electrostatically bound PIP5 kinases that are required to replenish PI(4,5)P2. As PI(4,5)P2 decreased, actin was depleted from the membrane, allowing secretion. Forced localization of PIP5Kβ across the synapse prevented actin depletion, blocking both centrosome docking and secretion. Thus, PIP5Ks act as molecular sensors of TCR activation, controlling actin recruitment across the synapse, ensuring exquisite co-ordination between TCR signaling and CTL secretion. Immune synapse formation triggers rapid changes in the membrane composition and charge PIP5K is a molecular sensor of TCR activation and is rapidly depleted at the synapse PIP5K distribution controls actin recruitment across the immune synapse Membrane specialization controls accessibility for centrosome docking and secretion
Collapse
Affiliation(s)
- Christian M Gawden-Bone
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Gordon L Frazer
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Arianne C Richard
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK; Cancer Research UK Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0RE, UK
| | - Claire Y Ma
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Katharina Strege
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| |
Collapse
|
43
|
Hackelberg S, Oliver D. Metabotropic Acetylcholine and Glutamate Receptors Mediate PI(4,5)P 2 Depletion and Oscillations in Hippocampal CA1 Pyramidal Neurons in situ. Sci Rep 2018; 8:12987. [PMID: 30154490 PMCID: PMC6113233 DOI: 10.1038/s41598-018-31322-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/17/2018] [Indexed: 01/24/2023] Open
Abstract
The sensitivity of many ion channels to phosphatidylinositol-4,5-bisphosphate (PIP2) levels in the cell membrane suggests that PIP2 fluctuations are important and general signals modulating neuronal excitability. Yet the PIP2 dynamics of central neurons in their native environment remained largely unexplored. Here, we examined the behavior of PIP2 concentrations in response to activation of Gq-coupled neurotransmitter receptors in rat CA1 hippocampal neurons in situ in acute brain slices. Confocal microscopy of the PIP2-selective molecular sensors tubbyCT-GFP and PLCδ1-PH-GFP showed that pharmacological activation of muscarinic acetylcholine (mAChR) or group I metabotropic glutamate (mGluRI) receptors induces transient depletion of PIP2 in the soma as well as in the dendritic tree. The observed PIP2 dynamics were receptor-specific, with mAChR activation inducing stronger PIP2 depletion than mGluRI, whereas agonists of other Gαq-coupled receptors expressed in CA1 neurons did not induce measureable PIP2 depletion. Furthermore, the data show for the first time neuronal receptor-induced oscillations of membrane PIP2 concentrations. Oscillatory behavior indicated that neurons can rapidly restore PIP2 levels during persistent activation of Gq and PLC. Electrophysiological responses to receptor activation resembled PIP2 dynamics in terms of time course and receptor specificity. Our findings support a physiological function of PIP2 in regulating electrical activity.
Collapse
Affiliation(s)
- Sandra Hackelberg
- Institute of Physiology and Pathophysiology, Philipps University, 35037, Marburg, Germany
- The Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Dominik Oliver
- Institute of Physiology and Pathophysiology, Philipps University, 35037, Marburg, Germany.
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps University, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), Marburg and Giessen, Germany.
| |
Collapse
|
44
|
Leitner MG, Thallmair V, Wilke BU, Neubert V, Kronimus Y, Halaszovich CR, Oliver D. The N-terminal homology (ENTH) domain of Epsin 1 is a sensitive reporter of physiological PI(4,5)P 2 dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:433-442. [PMID: 30670192 DOI: 10.1016/j.bbalip.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/18/2018] [Accepted: 08/04/2018] [Indexed: 11/15/2022]
Abstract
Phospholipase Cβ (PLCβ)-induced depletion of phosphatidylinositol-(4,5)-bisphosphate (PI(4,5)P2) transduces a plethora of signals into cellular responses. Importance and diversity of PI(4,5)P2-dependent processes led to strong need for biosensors of physiological PI(4,5)P2 dynamics applicable in live-cell experiments. Membrane PI(4,5)P2 can be monitored with fluorescently-labelled phosphoinositide (PI) binding domains that associate to the membrane depending on PI(4,5)P2 levels. The pleckstrin homology domain of PLCδ1 (PLCδ1-PH) and the C-terminus of tubby protein (tubbyCT) are two such sensors widely used to study PI(4,5)P2 signaling. However, certain limitations apply to both: PLCδ1-PH binds cytoplasmic inositol-1,4,5-trisphosphate (IP3) produced from PI(4,5)P2 through PLCβ, and tubbyCT responses do not faithfully report on PLCβ-dependent PI(4,5)P2 dynamics. In searching for an improved biosensor, we fused N-terminal homology domain of Epsin1 (ENTH) to GFP and examined use of this construct as genetically-encoded biosensor for PI(4,5)P2 dynamics in living cells. We utilized recombinant tools to manipulate PI or Gq protein-coupled receptors (GqPCR) to stimulate PLCβ signaling and characterized PI binding properties of ENTH-GFP with total internal reflection (TIRF) and confocal microscopy. ENTH-GFP specifically recognized membrane PI(4,5)P2 without interacting with IP3, as demonstrated by dialysis of cells with the messenger through a patch pipette. Utilizing Ci-VSP to titrate PI(4,5)P2 levels, we found that ENTH-GFP had low PI(4,5)P2 affinity. Accordingly, ENTH-GFP was highly sensitive to PLCβ-dependent PI(4,5)P2 depletion, and in contrast to PLCδ1-PH, overexpression of ENTH-GFP did not attenuate GqPCR signaling. Taken together, ENTH-GFP detects minute changes of PI(4,5)P2 levels and provides an important complementation of experimentally useful reporters of PI(4,5)P2 dynamics in physiological pathways.
Collapse
Affiliation(s)
- Michael G Leitner
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020 Innsbruck, Austria; Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany.
| | - Veronika Thallmair
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Bettina U Wilke
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Valentin Neubert
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Yannick Kronimus
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Christian R Halaszovich
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, 35037 Marburg, Germany; DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University, Germany; Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Germany
| |
Collapse
|
45
|
Chintaluri K, Goulden BD, Celmenza C, Saffi G, Miraglia E, Hammond GRV, Botelho RJ. The PH domain from the Toxoplasma gondii PH-containing protein-1 (TgPH1) serves as an ectopic reporter of phosphatidylinositol 3-phosphate in mammalian cells. PLoS One 2018; 13:e0198454. [PMID: 29870544 PMCID: PMC5988325 DOI: 10.1371/journal.pone.0198454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
Phosphoinositide (PtdInsP) lipids recruit effector proteins to membranes to mediate a variety of functions including signal transduction and membrane trafficking. Each PtdInsP binds to a specific set of effectors through characteristic protein domains such as the PH, FYVE and PX domains. Domains with high affinity for a single PtdInsP species are useful as probes to visualize the distribution and dynamics of that PtdInsP. The endolysosomal system is governed by two primary PtdInsPs: phosphatidylinositol 3-phosphate [PtdIns(3)P] and phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2], which are thought to localize and control early endosomes and lysosomes/late endosomes, respectively. While PtdIns(3)P has been analysed with mammalian-derived PX and FYVE domains, PtdIns(3,5)P2 indicators remain controversial. Thus, complementary probes against these PtdInsPs are needed, including those originating from non-mammalian proteins. Here, we characterized in mammalian cells the dynamics of the PH domain from PH-containing protein-1 from the parasite Toxoplasma gondii (TgPH1), which was previously shown to bind PtdIns(3,5)P2 in vitro. However, we show that TgPH1 retains membrane-binding in PIKfyve-inhibited cells, suggesting that TgPH1 is not a viable PtdIns(3,5)P2 marker in mammalian cells. Instead, PtdIns(3)P depletion using pharmacological and enzyme-based assays dissociated TgPH1 from membranes. Indeed, TgPH1 co-localized with Rab5-positive early endosomes. In addition, TgPH1 co-localized and behaved similarly to the PX domain of p40phox and FYVE domain of EEA1, which are commonly used as PtdIns(3)P indicators. Collectively, TgPH1 offers a complementary reporter for PtdIns(3)P derived from a non-mammalian protein and that is distinct from commonly employed PX and FYVE domain-based probes.
Collapse
Affiliation(s)
- Krishna Chintaluri
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
| | - Brady D. Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Camilyn Celmenza
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Golam Saffi
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
| | - Emily Miraglia
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
| | - Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Roberto J. Botelho
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
46
|
Del Vecchio K, Stahelin RV. Investigation of the phosphatidylserine binding properties of the lipid biosensor, Lactadherin C2 (LactC2), in different membrane environments. J Bioenerg Biomembr 2018; 50:1-10. [PMID: 29426977 DOI: 10.1007/s10863-018-9745-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/31/2018] [Indexed: 01/07/2023]
Abstract
Lipid biosensors are robust tools used in both in vitro and in vivo applications of lipid imaging and lipid detection. Lactadherin C2 (LactC2) was described in 2000 as being a potent and specific sensor for phosphatidylserine (PS) (Andersen et al. Biochemistry 39:6200-6206, 2000). PS is an anionic phospholipid enriched in the inner leaflet of the plasma membrane and has paramount roles in apoptosis, cells signaling, and autophagy. The myriad roles PS plays in membrane dynamics make monitoring PS levels and function an important endeavor. LactC2 has functioned as a tantamount PS biosensor namely in the field of cellular imaging. While PS specificity and high affinity of LactC2 for PS containing membranes has been well established, much less is known regarding LactC2 selectivity for subcellular pools of PS or PS within different membrane environments (e.g., in the presence of cholesterol). Thus, there has been a lack of studies that have compared LactC2 PS sensitivity based upon the acyl chain length and saturation or the presence of other host lipids such as cholesterol. Here, we use surface plasmon resonance as a label-free method to quantitatively assess the apparent binding affinity of LactC2 for membranes containing PS with different acyl chains, different fluidity, as well as representative lipid vesicle mimetics of cellular membranes. Results demonstrate that LactC2 is an unbiased sensor for PS, and can sensitively interact with membranes containing PS with different acyl chain saturation and interact with PS species in a cholesterol-independent manner.
Collapse
Affiliation(s)
- Kathryn Del Vecchio
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Robert V Stahelin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA. .,Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
47
|
Choy CH, Han BK, Botelho RJ. Phosphoinositide Diversity, Distribution, and Effector Function: Stepping Out of the Box. Bioessays 2017; 39. [PMID: 28977683 DOI: 10.1002/bies.201700121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/31/2017] [Indexed: 12/26/2022]
Abstract
Phosphoinositides (PtdInsPs) modulate a plethora of functions including signal transduction and membrane trafficking. PtdInsPs are thought to consist of seven interconvertible species that localize to a specific organelle, to which they recruit a set of cognate effector proteins. Here, in reviewing the literature, we argue that this model needs revision. First, PtdInsPs can carry a variety of acyl chains, greatly boosting their molecular diversity. Second, PtdInsPs are more promiscuous in their localization than is usually acknowledged. Third, PtdInsP interconversion is likely achieved through kinase-phosphatase enzyme complexes that coordinate their activities and channel substrates without affecting bulk substrate population. Additionally, we contend that despite hundreds of PtdInsP effectors, our attention is biased toward few proteins. Lastly, we recognize that PtdInsPs can act to nucleate coincidence detection at the effector level, as in PDK1 and Akt. Overall, better integrated models of PtdInsP regulation and function are not only possible but needed.
Collapse
Affiliation(s)
- Christopher H Choy
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada M5B2K3.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada M5B2K3
| | - Bong-Kwan Han
- The Intelligent Synthetic Biology Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Roberto J Botelho
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada M5B2K3.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada M5B2K3
| |
Collapse
|
48
|
Cortical actin recovery at the immunological synapse leads to termination of lytic granule secretion in cytotoxic T lymphocytes. Proc Natl Acad Sci U S A 2017; 114:E6585-E6594. [PMID: 28716933 DOI: 10.1073/pnas.1710751114] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) eliminate virally infected cells through directed secretion of specialized lytic granules. Because a single CTL can kill multiple targets, degranulation must be tightly regulated. However, how CTLs regulate the termination of granule secretion remains unclear. Previous work demonstrated that centralized actin reduction at the immune synapse precedes degranulation. Using a combination of live confocal, total internal reflection fluorescence, and superresolution microscopy, we now show that, after granule fusion, actin recovers at the synapse and no further secretion is observed. Depolymerization of actin led to resumed granule secretion, suggesting that recovered actin acts as a barrier preventing sustained degranulation. Furthermore, RAB27a-deficient CTLs, which do not secrete cytotoxic granules, failed to recover actin at the synapse, suggesting that RAB27a-mediated granule secretion is required for actin recovery. Finally, we show that both actin clearance and recovery correlated with synaptic phosphatidylinositol 4,5-bisphosphate (PIP2) and that alterations in PIP2 at the immunological synapse regulate cortical actin in CTLs, providing a potential mechanism through which CTLs control cortical actin density. Our work provides insight into actin-related mechanisms regulating CTL secretion that may facilitate serial killing during immune responses.
Collapse
|
49
|
Shi J, Miralles F, Kinet JP, Birnbaumer L, Large WA, Albert AP. Evidence that Orai1 does not contribute to store-operated TRPC1 channels in vascular smooth muscle cells. Channels (Austin) 2017; 11:329-339. [PMID: 28301277 PMCID: PMC5555289 DOI: 10.1080/19336950.2017.1303025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ca2+-permeable store-operated channels (SOCs) mediate Ca2+ entry pathways which are involved in many cellular functions such as contraction, growth, and proliferation. Prototypical SOCs are formed of Orai1 proteins and are activated by the endo/sarcoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1). There is considerable debate about whether canonical transient receptor potential 1 (TRPC1) proteins also form store-operated channels (SOCs), and if they do, is Orai1 involved. We recently showed that stimulation of TRPC1-based SOCs involves store depletion inducing STIM1-evoked Gαq/PLCβ1 activity in contractile vascular smooth muscle cells (VSMCs). Therefore the present work investigates the role of Orai1 in activation of TRPC1-based SOCs in freshly isolated mesenteric artery VSMCs from wild-type (WT) and Orai1−/− mice. Store-operated whole-cell and single channel currents recorded from WT and Orai1−/− VSMCs had similar properties, with relatively linear current-voltage relationships, reversal potentials of about +20mV, unitary conductances of about 2pS, and inhibition by anti-TRPC1 and anti-STIM1 antibodies. In Orai1−/− VSMCs, store depletion induced PLCβ1 activity measured with the fluorescent phosphatidylinositol 4,5-bisphosphate/inositol 1,4,5-trisphosphate biosensor GFP-PLCδ1-PH, which was prevented by knockdown of STIM1. In addition, in Orai1−/− VSMCs, store depletion induced translocation of STIM1 from within the cell to the plasma membrane where it formed STIM1-TRPC1 interactions at discrete puncta-like sites. These findings indicate that activation of TRPC1-based SOCs through a STIM1-activated PLCβ1 pathway are likely to occur independently of Orai1 proteins, providing evidence that TRPC1 channels form genuine SOCs in VSMCs with a contractile phenotype.
Collapse
Affiliation(s)
- Jian Shi
- a Institute of Cardiovascular & Metabolic Medicine, School of Medicine , University of Leeds , Leeds , UK
| | - Francesc Miralles
- b Vascular Biology Research Centre, Institute of Molecular & Clinical Sciences Research Institute , St. George's, University of London , Cranmer Terrace, London , UK.,c Institute of Medical & Biomedical Education, St. George's , University of London , Cranmer Terrace, London , UK
| | - Jean-Pierre Kinet
- d Laboratory of Allergy and Immunology, Department of Pathology, Beth Israel Deaconess Medical Center , Harvard Medical School , Boston , MA , USA
| | - Lutz Birnbaumer
- e Laboratory of Neurobiology , National Institute of Environmental Health Sciences , Research Triangle Park, NC , USA.,f Institute of Biomedical Research (BIOMED) , Catholic University of Argentina , Buenos Aires , Argentina
| | - William A Large
- b Vascular Biology Research Centre, Institute of Molecular & Clinical Sciences Research Institute , St. George's, University of London , Cranmer Terrace, London , UK
| | - Anthony P Albert
- b Vascular Biology Research Centre, Institute of Molecular & Clinical Sciences Research Institute , St. George's, University of London , Cranmer Terrace, London , UK
| |
Collapse
|
50
|
Shi J, Miralles F, Birnbaumer L, Large WA, Albert AP. Store-operated interactions between plasmalemmal STIM1 and TRPC1 proteins stimulate PLCβ1 to induce TRPC1 channel activation in vascular smooth muscle cells. J Physiol 2017; 595:1039-1058. [PMID: 27753095 PMCID: PMC5309361 DOI: 10.1113/jp273302] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/13/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Depletion of Ca2+ stores activates store-operated channels (SOCs), which mediate Ca2+ entry pathways that regulate cellular processes such as contraction, proliferation and gene expression. In vascular smooth muscle cells (VSMCs), stimulation of SOCs composed of canonical transient receptor potential channel 1 (TRPC1) proteins requires G protein α q subunit (Gαq)/phospholipase C (PLC)β1/protein kinase C (PKC) activity. We studied the role of stromal interaction molecule 1 (STIM1) in coupling store depletion to this activation pathway using patch clamp recording, GFP-PLCδ1-PH imaging and co-localization techniques. Store-operated TRPC1 channel and PLCβ1 activities were inhibited by STIM1 short hairpin RNA (shRNA) and absent in TRPC1-/- cells, and store-operated PKC phosphorylation of TRPC1 was inhibited by STIM1 shRNA. Store depletion induced interactions between STIM1 and TRPC1, Gαq and PLCβ1, which required STIM1 and TRPC1. Similar effects were produced with noradrenaline. These findings identify a new activation mechanism of TRPC1-based SOCs in VSMCs, and a novel role for STIM1, where store-operated STIM1-TRPC1 interactions stimulate Gαq/PLCβ1/PKC activity to induce channel gating. ABSTRACT In vascular smooth muscle cells (VSMCs), stimulation of canonical transient receptor potential channel 1 (TRPC1) protein-based store-operated channels (SOCs) mediates Ca2+ entry pathways that regulate contractility, proliferation and migration. It is therefore important to understand how these channels are activated. Studies have shown that stimulation of TRPC1-based SOCs requires G protein α q subunit (Gαq)/phospholipase C (PLC)β1 activities and protein kinase C (PKC) phosphorylation, although it is unclear how store depletion stimulates this gating pathway. The present study examines this issue by focusing on the role of stromal interaction molecule 1 (STIM1), an endo/sarcoplasmic reticulum Ca2+ sensor. Store-operated TRPC1 channel activity was inhibited by TRPC1 and STIM1 antibodies and STIM1 short hairpin RNA (shRNA) in wild-type VSMCs, and was absent in TRPC1-/- VSMCs. Store-operated PKC phosphorylation of TRPC1 was reduced by knockdown of STIM1. Moreover, store-operated PLCβ1 activity measured with the fluorescent phosphatidylinositol 4,5-bisphosphate/inositol 1,4,5-trisphosphate biosensor GFP-PLCδ1-PH was reduced by STIM1 shRNA and absent in TRPC1-/- cells. Immunocytochemistry, co-immunoprecipitation and proximity ligation assays revealed that store depletion activated STIM1 translocation from within the cell to the plasma membrane (PM) where it formed STIM1-TRPC1 complexes, which then associated with Gαq and PLCβ1. Noradrenaline also evoked TRPC1 channel activity and associations between TRPC1, STIM1, Gαq and PLCβ1, which were inhibited by STIM1 knockdown. Effects of N-terminal and C-terminal STIM1 antibodies on TRPC1-based SOCs and STIM1 staining suggest that channel activation may involve insertion of STIM1 into the PM. The findings of the present study identify a new activation mechanism of TRPC1-based SOCs in VSMCs, and a novel role for STIM1, in which store-operated STIM1-TRPC1 interactions stimulate PLCβ1 activity to induce PKC phosphorylation of TRPC1 and channel gating.
Collapse
Affiliation(s)
- Jian Shi
- Vascular Biology Research CentreMolecular & Clinical Sciences Research Institute
| | - Francesc Miralles
- Vascular Biology Research CentreMolecular & Clinical Sciences Research Institute
- Institute of Medical & Biomedical EducationSt George'sUniversity of LondonLondonUK
| | - Lutz Birnbaumer
- Neurobiology LaboratoryNational Institute of Environmental Health SciencesResearch Triangle ParkNCUSA
- Institute of Biomedical Research (BIOMED)School of Medical SciencesCatholic University of ArgentinaBuenos AiresArgentina
| | - William A. Large
- Vascular Biology Research CentreMolecular & Clinical Sciences Research Institute
| | - Anthony P. Albert
- Vascular Biology Research CentreMolecular & Clinical Sciences Research Institute
| |
Collapse
|