1
|
van Zanten TS, S GP, Mayor S. Quantitative fluorescence emission anisotropy microscopy for implementing homo-fluorescence resonance energy transfer measurements in living cells. Mol Biol Cell 2023; 34:tp1. [PMID: 37144969 DOI: 10.1091/mbc.e22-09-0446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
Quantitative fluorescence emission anisotropy microscopy reveals the organization of fluorescently labeled cellular components and allows their characterization in terms of changes in either rotational diffusion or homo-Förster's energy transfer characteristics in living cells. These properties provide insights into molecular organization, such as orientation, confinement, and oligomerization in situ. Here we elucidate how quantitative measurements of anisotropy using multiple microscope systems may be made by bringing out the main parameters that influence the quantification of fluorescence emission anisotropy. We focus on a variety of parameters that contribute to errors associated with the measurement of emission anisotropy in a microscope. These include the requirement for adequate photon counts for the necessary discrimination of anisotropy values, the influence of extinction ratios of the illumination source, the detector system, the role of numerical aperture, and excitation wavelength. All these parameters also affect the ability to capture the dynamic range of emission anisotropy necessary for quantifying its reduction due to homo-FRET and other processes. Finally, we provide easily implementable tests to assess whether homo-FRET is a cause for the observed emission depolarization.
Collapse
Affiliation(s)
- Thomas S van Zanten
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| | - Greeshma Pradeep S
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| | - Satyajit Mayor
- Cell Biology Group, National Centre for Biological Sciences, UAS-GKVK Campus, Tata Institute for Fundamental Research, Bangalore 560065, India
| |
Collapse
|
2
|
Sanfeliu-Cerdán N, Lin LC, Dunn AR, Goodman MB, Krieg M. Visualizing Neurons Under Tension In Vivo with Optogenetic Molecular Force Sensors. Methods Mol Biol 2023; 2600:239-266. [PMID: 36587102 DOI: 10.1007/978-1-0716-2851-5_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The visualization of mechanical stress distribution in specific molecular networks within a living and physiologically active cell or animal remains a formidable challenge in mechanobiology. The advent of fluorescence-resonance energy transfer (FRET)-based molecular tension sensors overcame a significant hurdle that now enables us to address previously technically limited questions. Here, we describe a method that uses genetically encoded FRET tension sensors to visualize the mechanics of cytoskeletal networks in neurons of living animals with sensitized emission FRET and confocal scanning light microscopy. This method uses noninvasive immobilization of living animals to image neuronal β-spectrin cytoskeleton at the diffraction limit, and leverages multiple imaging controls to verify and underline the quality of the measurements. In combination with a semiautomated machine-vision algorithm to identify and trace individual neurites, our analysis performs simultaneous calculation of FRET efficiencies and visualizes statistical uncertainty on a pixel by pixel basis. Our approach is not limited to genetically encoded spectrin tension sensors, but can also be used for any kind of ratiometric imaging in neuronal cells both in vivo and in vitro.
Collapse
Affiliation(s)
- Neus Sanfeliu-Cerdán
- Neurophotonics and Mechanical Systems Biology, ICFO, Institut de Ciències Fotòniques, ICFO, Castelldefels, Spain
| | - Li-Chun Lin
- Neurophotonics and Mechanical Systems Biology, ICFO, Institut de Ciències Fotòniques, ICFO, Castelldefels, Spain
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Miriam B Goodman
- Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Michael Krieg
- Neurophotonics and Mechanical Systems Biology, ICFO, Institut de Ciències Fotòniques, ICFO, Castelldefels, Spain.
| |
Collapse
|
3
|
Abstract
This unit describes the basic principles of Förster resonance energy transfer (FRET). Beginning with a brief summary of the history of FRET applications, the theory of FRET is introduced in detail using figures to explain all the important parameters of the FRET process. After listing various approaches for measuring FRET efficiency, several pieces of advice are given on choosing the appropriate instrumentation. The unit concludes with a discussion of the limitations of FRET measurements followed by a few examples of the latest FRET applications, including new developments such as spectral flow cytometric FRET, single-molecule FRET, and combinations of FRET with super-resolution or lifetime imaging microscopy and with molecular dynamics simulations. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Ágnes Szabó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
4
|
Dai G. Neuronal KCNQ2/3 channels are recruited to lipid raft microdomains by palmitoylation of BACE1. J Gen Physiol 2022; 154:213033. [PMID: 35201266 PMCID: PMC8876601 DOI: 10.1085/jgp.202112888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
β-Secretase 1 (β-site amyloid precursor protein [APP]-cleaving enzyme 1, BACE1) plays a crucial role in the amyloidogenesis of Alzheimer’s disease (AD). BACE1 was also discovered to act like an auxiliary subunit to modulate neuronal KCNQ2/3 channels independently of its proteolytic function. BACE1 is palmitoylated at its carboxyl-terminal region, which brings BACE1 to ordered, cholesterol-rich membrane microdomains (lipid rafts). However, the physiological consequences of this specific localization of BACE1 remain elusive. Using spectral Förster resonance energy transfer (FRET), BACE1 and KCNQ2/3 channels were confirmed to form a signaling complex, a phenomenon that was relatively independent of the palmitoylation of BACE1. Nevertheless, palmitoylation of BACE1 was required for recruitment of KCNQ2/3 channels to lipid-raft domains. Two fluorescent probes, designated L10 and S15, were used to label lipid-raft and non-raft domains of the plasma membrane, respectively. Coexpressing BACE1 substantially elevated FRET between L10 and KCNQ2/3, whereas the BACE1-4C/A quadruple mutation failed to produce this effect. In contrast, BACE1 had no significant effect on FRET between S15 probes and KCNQ2/3 channels. A reduction of BACE1-dependent FRET between raft-targeting L10 probes and KCNQ2/3 channels by applying the cholesterol-extracting reagent methyl-β-cyclodextrin (MβCD), raft-disrupting general anesthetics, or pharmacological inhibitors of palmitoylation, all supported the hypothesis of the palmitoylation-dependent and raft-specific localization of KCNQ2/3 channels. Furthermore, mutating the four carboxyl-terminal cysteines (4C/A) of BACE1 abolished the BACE1-dependent increase of FRET between KCNQ2/3 and the lipid raft–specific protein caveolin 1. Taking these data collectively, we propose that the AD-related protein BACE1 underlies the localization of a neuronal potassium channel.
Collapse
Affiliation(s)
- Gucan Dai
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
5
|
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
|
6
|
Liao P, Zang S, Wu T, Jin H, Wang W, Huang J, Tang BZ, Yan Y. Generating circularly polarized luminescence from clusterization-triggered emission using solid phase molecular self-assembly. Nat Commun 2021; 12:5496. [PMID: 34535652 PMCID: PMC8448880 DOI: 10.1038/s41467-021-25789-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
Purely-organic clusterization-triggered emission (CTE) has displayed promising abilities in bioimaging, chemical sensing, and multicolor luminescence. However, it remains absent in the field of circularly polarized luminescence (CPL) due to the difficulties in well-aligning the nonconventional luminogens. We report a case of CPL generated with CTE using the solid phase molecular self-assembly (SPMSA) of poly-L-lysine (PLL) and oleate ion (OL), that is, the macroscopic CPL supramolecular film self-assembled by the electrostatic complex of PLL/OL under mechanical pressure. Well-defined interface charge distribution, given by lamellar mesophases of OL ions, forces the PLL chains to fold regularly as a requirement of optimal electrostatic interactions. Further facilitated by hydrogen bonding, the through-space conjugation (TSC) of orderly aligned electron-rich O and N atoms leads to CTE-based CPL, which is capable of transferring energy to an acceptor via a Förster resonance energy transfer (FRET) process, making it possible to develop environmentally friendly and economic CPL from sustainable and renewable materials.
Collapse
Affiliation(s)
- Peilong Liao
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Shihao Zang
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Tongyue Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Hongjun Jin
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Wenkai Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Jianbin Huang
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Ben Zhong Tang
- Shenzhen Institute of Molecular Aggregate Science and Engineering, School of Science and Engineering, The Chinese University of Hong Kong, 2001 Longxiang Boulevard, Longgang, Shenzhen, Guangdong, 518172, China.
| | - Yun Yan
- Beijing National Laboratory for Molecular Sciences (BNLMS), College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
7
|
Abstract
Lateral organization in the plane of the plasma membrane is an important driver of biological processes. The past dozen years have seen increasing experimental support for the notion that lipid organization plays an important role in modulating this heterogeneity. Various biophysical mechanisms rooted in the concept of liquid-liquid phase separation have been proposed to explain diverse experimental observations of heterogeneity in model and cell membranes with distinct but overlapping applicability. In this review, we focus on the evidence for and the consequences of the hypothesis that the plasma membrane is poised near an equilibrium miscibility critical point. Critical phenomena explain certain features of the heterogeneity observed in cells and model systems but also go beyond heterogeneity to predict other interesting phenomena, including responses to perturbations in membrane composition.
Collapse
Affiliation(s)
- Thomas R Shaw
- Program in Applied Physics, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Subhadip Ghosh
- Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Sarah L Veatch
- Program in Applied Physics, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Physics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
8
|
Interactions of different lipoproteins with supported phospholipid raft membrane (SPRM) patterns to understand similar in-vivo processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183535. [PMID: 33358851 DOI: 10.1016/j.bbamem.2020.183535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 11/21/2022]
Abstract
To better understand how lipoproteins interact and enter endothelium and participate in cellular processes, we investigated preferential lipid partitioning of triglyceride rich lipoproteins (TGRL), chylomicrons (CM), low density lipoproteins (LDL), very low density lipoproteins (VLDL) and their lipolysis products using supported phospholipid raft membrane (SPRM) patterns. We prepared SPRM patterns with Texas red labeled phospholipid patterns and Marina blue labeled raft patterns and added Atto-520 labeled lipoproteins (TGRL, CM, VLDL, LDL) and their lipolysis products in separate experiments and characterized these interactions using fluorescence microscopy. We observed that VLDL and LDL preferentially interacted with raft patterns. In contrast the TGRL and lipolysed products of TGRL interacted with both the patterns, slightly elevated preference for raft patterns and CM and its lipolysis products showed greater affinity to phospholipid patterns. The clear preference of VLDL and LDL for raft patterns suggests that these lipoproteins associate with cholesterol and sphingomyelin rich lipid micro-domains during their early interactions with endothelial cells, leading to atherosclerosis.
Collapse
|
9
|
Scott HL, Baker JR, Frederick AJ, Kennison KB, Mendes K, Heberle FA. FRET from phase-separated vesicles: An analytical solution for a spherical geometry. Chem Phys Lipids 2020; 233:104982. [PMID: 33065119 DOI: 10.1016/j.chemphyslip.2020.104982] [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] [Received: 06/22/2020] [Revised: 09/20/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
Förster resonance energy transfer (FRET) is a powerful tool for investigating heterogeneity in lipid bilayers. In model membrane studies, samples are frequently unilamellar vesicles with diameters of 20-200 nm. It is well-known that FRET efficiency is insensitive to vesicle curvature in uniformly mixed lipid bilayers, and consequently theoretical models for FRET typically assume a planar geometry. Here, we use a spherical harmonic expansion of the acceptor surface density to derive an analytical solution for FRET between donor and acceptor molecules distributed on the surface of a sphere. We find excellent agreement between FRET predicted from the model and FRET calculated from corresponding Monte Carlo simulations, thus validating the model. An extension of the model to the case of a non-uniform acceptor surface density (i.e., a phase-separated vesicle) reveals that FRET efficiency depends on vesicle size when acceptors partition between the coexisting phases, and approaches the efficiency of a uniformly mixed bilayer as the vesicle size decreases. We show that this is an indirect effect of constrained domain size, rather than an intrinsic effect of vesicle curvature. Surprisingly, the theoretical predictions were not borne out in experiments: we did not observe a statistically significant change in FRET efficiency in phase-separated vesicles as a function of vesicle size. We discuss factors that likely mask the vesicle size effect in extruded samples.
Collapse
Affiliation(s)
- Haden L Scott
- Center for Environmental Biotechnology, University of Tennessee, Knoxville, TN 37920, United States; Shull Wollan Center, Oak Ridge National Laboratory, Oak Ridge, TN 37831, United States; Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - James R Baker
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Aaron J Frederick
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Kristen B Kennison
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Kevin Mendes
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Frederick A Heberle
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States.
| |
Collapse
|
10
|
Almada E, Pariani A, Rivabella Maknis T, Hidalgo F, Vena R, Favre C, Larocca MC. AKAP350 enables p150glued /EB1 interaction at the spindle poles. Biochimie 2020; 177:127-131. [PMID: 32841682 DOI: 10.1016/j.biochi.2020.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 11/28/2022]
Abstract
A-kinase anchoring protein 350 (AKAP350) is a centrosomal/Golgi scaffold protein, critical for the regulation of microtubule dynamics. AKAP350 recruits end-binding protein 1 (EB1) to the centrosome in mitotic cells, ensuring proper spindle orientation in epithelial cells. AKAP350 also interacts with p150glued, the main component of the dynactin complex. In the present work, we found that AKAP350 localized p150glued to the spindle poles, facilitating p150glued/EB1 interaction at these structures. Our results further showed that the decrease in AKAP350 expression reduced p150glued localization at astral microtubules and impaired the elongation of astral microtubules during anaphase. Overall, this study provides mechanistic data on how microtubule regulatory proteins gather to define microtubule dynamics in mitotic cells.
Collapse
Affiliation(s)
- Evangelina Almada
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina
| | - Alejandro Pariani
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina
| | - Tomás Rivabella Maknis
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina
| | - Florencia Hidalgo
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina
| | - Rodrigo Vena
- Instituto de Biología Molecular y Cellular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | - Cristián Favre
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina
| | - M Cecilia Larocca
- Instituto de Fisiología Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), 2000, Rosario, Argentina.
| |
Collapse
|
11
|
Szabó Á, Szendi-Szatmári T, Szöllősi J, Nagy P. Quo vadis FRET? Förster's method in the era of superresolution. Methods Appl Fluoresc 2020; 8:032003. [PMID: 32521530 DOI: 10.1088/2050-6120/ab9b72] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the theoretical foundations of Förster resonance energy transfer (FRET) were laid in the 1940s as part of the quantum physical revolution of the 20th century, it was only in the 1970s that it made its way to biology as a result of the availability of suitable measuring and labeling technologies. Thanks to its ease of application, FRET became widely used for studying molecular associations on the nanometer scale. The development of superresolution techniques at the turn of the millennium promised an unprecedented insight into the structure and function of molecular complexes. Without downplaying the significance of superresolution microscopies this review expresses our view that FRET is still a legitimate tool in the armamentarium of biologists for studying molecular associations since it offers distinct advantages and overcomes certain limitations of superresolution approaches.
Collapse
Affiliation(s)
- Ágnes Szabó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032 Debrecen, Hungary. MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032 Debrecen, Hungary
| | | | | | | |
Collapse
|
12
|
Balatskaya MN, Baglay AI, Rubtsov YP, Sharonov GV. Analysis of GPI-Anchored Receptor Distribution and Dynamics in Live Cells by Tag-mediated Enzymatic Labeling and FRET. Methods Protoc 2020; 3:mps3020033. [PMID: 32349461 PMCID: PMC7359698 DOI: 10.3390/mps3020033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/25/2020] [Accepted: 04/26/2020] [Indexed: 01/17/2023] Open
Abstract
The analysis of glycosylphosphatidylinositol (GPI)-anchored receptor distribution and dynamics in live cells is challenging, because their clusters exhibit subdiffraction-limited sizes and are highly dynamic. However, the cellular response depends on the GPI-anchored receptor clusters' distribution and dynamics. Here, we compare three approaches to GPI-anchored receptor labeling (with antibodies, fluorescent proteins, and enzymatically modified small peptide tags) and use several variants of Förster resonance energy transfer (FRET) detection by confocal microscopy and flow cytometry in order to obtain insight into the distribution and the ligand-induced dynamics of GPI-anchored receptors. We found that the enzyme-mediated site-specific fluorescence labeling of T-cadherin modified with a short peptide tag (12 residues in length) have several advantages over labeling by fluorescent proteins or antibodies, including (i) the minimized distortion of the protein's properties, (ii) the possibility to use a cell-impermeable fluorescent substrate that allows for selective labeling of surface-exposed proteins in live cells, and (iii) superior control of the donor to acceptor molar ratio. We successfully detected the FRET of GPI-anchored receptors, T-cadherin, and ephrin-A1, without ligands, and showed in real time that adiponectin induces stable T-cadherin cluster formation. In this paper (which is complementary to our recent research (Balatskaya et al., 2019)), we present the practical aspects of labeling and the heteroFRET measurements of GPI-anchored receptors to study their dynamics on a plasma membrane in live cells.
Collapse
Affiliation(s)
- Maria N. Balatskaya
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovskiy av. 27-1, 119192 Moscow, Russia; (A.I.B.); (Y.P.R.); (G.V.S.)
- Correspondence:
| | - Alexandra I. Baglay
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovskiy av. 27-1, 119192 Moscow, Russia; (A.I.B.); (Y.P.R.); (G.V.S.)
| | - Yury P. Rubtsov
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovskiy av. 27-1, 119192 Moscow, Russia; (A.I.B.); (Y.P.R.); (G.V.S.)
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry RAS, str. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - George V. Sharonov
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovskiy av. 27-1, 119192 Moscow, Russia; (A.I.B.); (Y.P.R.); (G.V.S.)
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry RAS, str. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Ostrovitianov str. 1, 117997 Moscow, Russia
- Laboratory of Genomics of Antitumor Adaptive Immunity, Privolzhsky Research Medical University, 10/1 Minin & Pozharsky sq., 603005 Nizhny Novgorod, Russia
| |
Collapse
|
13
|
Levental I, Levental KR, Heberle FA. Lipid Rafts: Controversies Resolved, Mysteries Remain. Trends Cell Biol 2020; 30:341-353. [PMID: 32302547 DOI: 10.1016/j.tcb.2020.01.009] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 01/08/2023]
Abstract
The lipid raft hypothesis postulates that lipid-lipid interactions can laterally organize biological membranes into domains of distinct structures, compositions, and functions. This proposal has in equal measure exhilarated and frustrated membrane research for decades. While the physicochemical principles underlying lipid-driven domains has been explored and is well understood, the existence and relevance of such domains in cells remains elusive, despite decades of research. Here, we review the conceptual underpinnings of the raft hypothesis and critically discuss the supporting and contradicting evidence in cells, focusing on why controversies about the composition, properties, and even the very existence of lipid rafts remain unresolved. Finally, we highlight several recent breakthroughs that may resolve existing controversies and suggest general approaches for moving beyond questions of the existence of rafts and towards understanding their physiological significance.
Collapse
Affiliation(s)
- Ilya Levental
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 70030, USA.
| | - Kandice R Levental
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 70030, USA
| | - Frederick A Heberle
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA; Shull Wollan Center, Oak Ridge National Laboratory, Oak Ridge, TN 33830, USA
| |
Collapse
|
14
|
Different spatiotemporal organization of GPI-anchored T-cadherin in response to low-density lipoprotein and adiponectin. Biochim Biophys Acta Gen Subj 2019; 1863:129414. [DOI: 10.1016/j.bbagen.2019.129414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 07/23/2019] [Accepted: 08/07/2019] [Indexed: 01/10/2023]
|
15
|
Tonucci FM, Almada E, Borini-Etichetti C, Pariani A, Hidalgo F, Rico MJ, Girardini J, Favre C, Goldenring JR, Menacho-Marquez M, Larocca MC. Identification of a CIP4 PKA phosphorylation site involved in the regulation of cancer cell invasiveness and metastasis. Cancer Lett 2019; 461:65-77. [DOI: 10.1016/j.canlet.2019.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 01/08/2023]
|
16
|
IL-2 receptors preassemble and signal in the ER/Golgi causing resistance to antiproliferative anti-IL-2Rα therapies. Proc Natl Acad Sci U S A 2019; 116:21120-21130. [PMID: 31570576 DOI: 10.1073/pnas.1901382116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interleukin-2 (IL-2) and IL-15 play pivotal roles in T cell activation, apoptosis, and survival, and are implicated in leukemias and autoimmune diseases. Their heterotrimeric receptors share their β- and γc-chains, but have distinct α-chains. Anti-IL-2Rα (daclizumab) therapy targeting cell surface-expressed receptor subunits to inhibit T cell proliferation has only brought limited success in adult T cell leukemia/lymphoma (ATL) and in multiple sclerosis. We asked whether IL-2R subunits could already preassemble and signal efficiently in the endoplasmic reticulum (ER) and the Golgi. A combination of daclizumab and anti-IL-2 efficiently blocked IL-2-induced proliferation of IL-2-dependent wild-type (WT) ATL cells but not cells transfected with IL-2, suggesting that in IL-2-producing cells signaling may already take place before receptors reach the cell surface. In the Golgi fraction isolated from IL-2-producing ATL cells, we detected by Western blot phosphorylated Jak1, Jak3, and a phosphotyrosine signal attributed to the γc-chain, which occurred at much lower levels in the Golgi of WT ATL cells. We expressed EGFP- and mCherry-tagged receptor chains in HeLa cells to study their assembly along the secretory pathway. Confocal microscopy, Förster resonance energy transfer, and imaging fluorescence cross-correlation spectroscopy analysis revealed partial colocalization and molecular association of IL-2 (and IL-15) receptor chains in the ER/Golgi, which became more complete in the plasma membrane, further confirming our hypothesis. Our results define a paradigm of intracellular autocrine signaling and may explain resistance to antagonistic antibody therapies targeting receptors at the cell surface.
Collapse
|
17
|
Nanoscale analysis reveals no domain formation of glycosylphosphatidylinositol-anchored protein SAG1 in the plasma membrane of living Toxoplasma gondii. Histochem Cell Biol 2019; 152:365-375. [PMID: 31542792 DOI: 10.1007/s00418-019-01814-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2019] [Indexed: 10/25/2022]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins typically localise to lipid rafts. GPI-anchored protein microdomains may be present in the plasma membrane; however, they have been studied using heterogeneously expressed GPI-anchored proteins, and the two-dimensional distributions of endogenous molecules in the plasma membrane are difficult to determine at the nanometre scale. Here, we used immunoelectron microscopy using a quick-freezing and freeze-fracture labelling (QF-FRL) method to examine the distribution of the endogenous GPI-anchored protein SAG1 in Toxoplasma gondii at the nanoscale. QF-FRL physically immobilised molecules in situ, minimising the possibility of artefactual perturbation. SAG1 labelling was observed in the exoplasmic, but not cytoplasmic, leaflets of T. gondii plasma membrane, whereas none was detected in any leaflet of the inner membrane complex. Point pattern analysis of SAG1 immunogold labelling revealed mostly random distribution in T. gondii plasma membrane. The present method obtains information on the molecular distribution of natively expressed GPI-anchored proteins and demonstrates that SAG1 in T. gondii does not form significant microdomains in the plasma membrane.
Collapse
|
18
|
Single Proteoliposome High-Content Analysis Reveals Differences in the Homo-Oligomerization of GPCRs. Biophys J 2019; 115:300-312. [PMID: 30021106 DOI: 10.1016/j.bpj.2018.05.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 11/23/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) control vital cellular signaling pathways. GPCR oligomerization is proposed to increase signaling diversity. However, many reports have arrived at disparate conclusions regarding the existence, stability, and stoichiometry of GPCR oligomers, partly because of cellular complexity and ensemble averaging of intrareconstitution heterogeneities that complicate the interpretation of oligomerization data. To overcome these limitations, we exploited fluorescence-microscopy-based high-content analysis of single proteoliposomes. This allowed multidimensional quantification of intrinsic monomer-monomer interactions of three class A GPCRs (β2-adrenergic receptor, cannabinoid receptor type 1, and opsin). Using a billion-fold less protein than conventional assays, we quantified oligomer stoichiometries, association constants, and the influence of two ligands and membrane curvature on oligomerization, revealing key similarities and differences for three GPCRs with decidedly different physiological functions. The assays introduced here will assist with the quantitative experimental observation of oligomerization for transmembrane proteins in general.
Collapse
|
19
|
Szendi-Szatmári T, Szabó Á, Szöllősi J, Nagy P. Reducing the Detrimental Effects of Saturation Phenomena in FRET Microscopy. Anal Chem 2019; 91:6378-6382. [DOI: 10.1021/acs.analchem.9b01504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tímea Szendi-Szatmári
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem Square 1, 4032 Debrecen, Hungary
| | - Ágnes Szabó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem Square 1, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem Square 1, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem Square 1, 4032 Debrecen, Hungary
| |
Collapse
|
20
|
Zong H, Wang X, Mu X, Wang J, Sun M. Plasmon-Enhanced Fluorescence Resonance Energy Transfer. CHEM REC 2019; 19:818-842. [PMID: 30716206 DOI: 10.1002/tcr.201800181] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/07/2019] [Indexed: 11/12/2022]
Abstract
In this review, we firstly introduce physical mechanism of fluorescence resonance energy transfer (FRET), the methods to measure FRET efficiency, and the applications of FRET. Secondly, we introduce the principle and applications of plasmon-enhanced fluorescence (PEF). Thirdly, we focused on the principle and applications of plasmon-enhanced FRET. This review can promote further understanding of FRET and PE-FRET.
Collapse
Affiliation(s)
- Huan Zong
- Computational Center for Property and Modification on Nanomaterials, College of Science, Liaoning Shihua University, Fushun, 113001, People's Republic of China.,School of Mathematics and Physics, Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Magneto-Photoelectrical Composite and Interface Science, University of Science and Technology Beijing, Beijing, 100083, People's Republic of China
| | - Xinxin Wang
- School of Mathematics and Physics, Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Magneto-Photoelectrical Composite and Interface Science, University of Science and Technology Beijing, Beijing, 100083, People's Republic of China
| | - Xijiao Mu
- School of Mathematics and Physics, Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Magneto-Photoelectrical Composite and Interface Science, University of Science and Technology Beijing, Beijing, 100083, People's Republic of China
| | - Jingang Wang
- Computational Center for Property and Modification on Nanomaterials, College of Science, Liaoning Shihua University, Fushun, 113001, People's Republic of China
| | - Mengtao Sun
- School of Mathematics and Physics, Beijing Advanced Innovation Center for Materials Genome Engineering, Beijing Key Laboratory for Magneto-Photoelectrical Composite and Interface Science, University of Science and Technology Beijing, Beijing, 100083, People's Republic of China
| |
Collapse
|
21
|
Susec M, Sencanski M, Glisic S, Veljkovic N, Pedersen C, Drinovec L, Stojan J, Nøhr J, Vrecl M. Functional characterization of β 2-adrenergic and insulin receptor heteromers. Neuropharmacology 2019; 152:78-89. [PMID: 30707913 DOI: 10.1016/j.neuropharm.2019.01.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/01/2019] [Accepted: 01/23/2019] [Indexed: 01/06/2023]
Abstract
This study aimed to functionally characterize β2-adrenergic (β2AR) and insulin receptor (IR) heteromers in regard to β-arrestin 2 (βarr2) recruitment and cAMP signaling and to examine the involvement of the cytoplasmic portion of the IR β chain in heteromerization with β2AR. Evidence for β2AR:IR:βarr2 complex formation and the specificity of the IR:βarr2 interaction was first provided by bioinfomatics analysis. Receptor-heteromer investigation technology (HIT) then provided functional evidence of β2AR:IR heterodimerization by showing isoproterenol-induced but not insulin-induced GFP2-βarr2 recruitment to the β2AR:IR complex; the IR:βarr2 interaction was found to only be constitutive. The constitutive IR:βarr2 BRET signal (BRETconst) was significantly smaller in cells coexpressing IR-RLuc8 and a GFP2-βarr2 1-185 mutant lacking the proposed IR binding domain. β2AR:IR heteromerization also influenced the pharmacological phenotype of β2AR, i.e., its efficacy in recruiting βarr2 and activating cAMP signaling. Evidence suggesting involvement of the cytoplasmic portion of the IR β chain in the interaction with β2AR was provided by BRET2 saturation and HIT assays using an IR 1-1271 stop mutant lacking the IR C-terminal tail region. For the complex consisting of IR 1-1271-RLuc8:β2AR-GFP2, saturation was not reached, most likely reflecting random collisions between IR 1-1271 and β2AR. Furthermore, in the HIT assay, no substantial agonist-induced increase in the BRET2 signal was detected that would be indicative of βarr2 recruitment to the IR 1-1271:β2AR heteromer. Complementary 3D visualization of β2AR:IR provided supporting evidence for stability of the heterotetramer complex and identified amino acid residues involved in β2AR:IR heteromerization. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Maja Susec
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Slovenia
| | - Milan Sencanski
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Sanja Glisic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Nevena Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Christina Pedersen
- Department of Incretin & Islet Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Luka Drinovec
- Department of Condensed Matter Physics, Jožef Stefan Institute, Slovenia
| | - Jurij Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jane Nøhr
- Department of Incretin & Islet Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Slovenia.
| |
Collapse
|
22
|
Ziegler CS, Bouchab L, Tramier M, Durand D, Fieschi F, Dupré-Crochet S, Mérola F, Nüße O, Erard M. Quantitative live-cell imaging and 3D modeling reveal critical functional features in the cytosolic complex of phagocyte NADPH oxidase. J Biol Chem 2019; 294:3824-3836. [PMID: 30630949 DOI: 10.1074/jbc.ra118.006864] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/08/2019] [Indexed: 01/18/2023] Open
Abstract
Phagocyte NADPH oxidase produces superoxide anions, a precursor of reactive oxygen species (ROS) critical for host responses to microbial infections. However, uncontrolled ROS production contributes to inflammation, making NADPH oxidase a major drug target. It consists of two membranous (Nox2 and p22phox) and three cytosolic subunits (p40phox, p47phox, and p67phox) that undergo structural changes during enzyme activation. Unraveling the interactions between these subunits and the resulting conformation of the complex could shed light on NADPH oxidase regulation and help identify inhibition sites. However, the structures and the interactions of flexible proteins comprising several well-structured domains connected by intrinsically disordered protein segments are difficult to investigate by conventional techniques such as X-ray crystallography, NMR, or cryo-EM. Here, we developed an analytical strategy based on FRET-fluorescence lifetime imaging (FLIM) and fluorescence cross-correlation spectroscopy (FCCS) to structurally and quantitatively characterize NADPH oxidase in live cells. We characterized the inter- and intramolecular interactions of its cytosolic subunits by elucidating their conformation, stoichiometry, interacting fraction, and affinities in live cells. Our results revealed that the three subunits have a 1:1:1 stoichiometry and that nearly 100% of them are present in complexes in living cells. Furthermore, combining FRET data with small-angle X-ray scattering (SAXS) models and published crystal structures of isolated domains and subunits, we built a 3D model of the entire cytosolic complex. The model disclosed an elongated complex containing a flexible hinge separating two domains ideally positioned at one end of the complex and critical for oxidase activation and interactions with membrane components.
Collapse
Affiliation(s)
- Cornelia S Ziegler
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France
| | - Leïla Bouchab
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France
| | - Marc Tramier
- the Université Rennes, CNRS, Institut de Génétique et Développement de Rennes - UMR 6290, BIOSIT - UMS 3480, F-35000 Rennes, France
| | - Dominique Durand
- the Institute for Integrative Biology of the Cell, CEA, CNRS UMR 9198, Université Paris-Sud, Université Paris-Saclay, 91190 Gif-sur-Yvette, France, and
| | - Franck Fieschi
- the Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Sophie Dupré-Crochet
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France
| | - Fabienne Mérola
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France
| | - Oliver Nüße
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France,
| | - Marie Erard
- From the Laboratoire de Chimie Physique, CNRS, Université Paris-Sud, Université Paris-Saclay, 91405, Orsay France,
| |
Collapse
|
23
|
Clayton AH. Fluorescence-based approaches for monitoring membrane receptor oligomerization. J Biosci 2018; 43:463-469. [PMID: 30002266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Membrane protein structures are highly under-represented relative to water-soluble protein structures in the protein databank. This is especially the case because membrane proteins represent more than 30% of proteins encoded in the human genome yet contribute to less than 10% of currently known structures (Torres et al. in Trends Biol Sci 28:137-144, 2003). Obtaining high-resolution structures of membrane proteins by traditional methods such as NMR and x-ray crystallography is challenging, because membrane proteins are difficult to solubilise, purify and crystallize. Consequently, development of methods to examine protein structure in situ is highly desirable. Fluorescence is highly sensitive to protein structure and dynamics (Lakowicz in Principles of fluorescence spectroscopy, Springer, New York, 2007). This is mainly because of the time a fluorescence probe molecule spends in the excited state. Judicious choice and placement of fluorescent molecule(s) within a protein(s) enables the experimentalist to obtain information at a specific site(s) in the protein (complex) of interest. Moreover, the inherent multi-dimensional nature of fluorescence signals across wavelength, orientation, space and time enables the design of experiments that give direct information on protein structure and dynamics in a biological setting. The purpose of this review is to introduce the reader to approaches to determine oligomeric state or quaternary structure at the cell membrane surface with the ultimate goal of linking the oligomeric state to the biological function. In the first section, we present a brief overview of available methods for determining oligomeric state and compare their advantages and disadvantages. In the second section, we highlight some of the methods developed in our laboratory to address contemporary questions in membrane protein oligomerization. In the third section, we outline our approach to determine the link between protein oligomerization and biological activity.
Collapse
Affiliation(s)
- Andrew Ha Clayton
- Cell Biophysics Laboratory, Centre for Micro-Photonics, Department of Physics and Astronomy, School of Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Australia,
| |
Collapse
|
24
|
Fluorescence-based approaches for monitoring membrane receptor oligomerization. J Biosci 2018. [DOI: 10.1007/s12038-018-9762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
25
|
Hooghoudt JO, Barroso M, Waagepetersen R. TOWARD BAYESIAN INFERENCE OF THE SPATIAL DISTRIBUTION OF PROTEINS FROM THREE-CUBE FÖRSTER RESONANCE ENERGY TRANSFER DATA. Ann Appl Stat 2018; 11:1711-1737. [PMID: 29861820 DOI: 10.1214/17-aoas1054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Förster resonance energy transfer (FRET) is a quantum-physical phenomenon where energy may be transferred from one molecule to a neighbor molecule if the molecules are close enough. Using fluorophore molecule marking of proteins in a cell, it is possible to measure in microscopic images to what extent FRET takes place between the fluorophores. This provides indirect information of the spatial distribution of the proteins. Questions of particular interest are whether (and if so to which extent) proteins of possibly different types interact or whether they appear independently of each other. In this paper we propose a new likelihood-based approach to statistical inference for FRET microscopic data. The likelihood function is obtained from a detailed modeling of the FRET data-generating mechanism conditional on a protein configuration. We next follow a Bayesian approach and introduce a spatial point process prior model for the protein configurations depending on hyperparameters quantifying the intensity of the point process. Posterior distributions are evaluated using Markov chain Monte Carlo. We propose to infer microscope-related parameters in an initial step from reference data without interaction between the proteins. The new methodology is applied to simulated and real datasets.
Collapse
|
26
|
Abstract
In the past decades, investigation of protein-protein interactions in situ in living or intact cells has gained expanding importance as structure/function relationships proposed from bulk biochemistry and molecular modeling experiments required confirmation at the cellular level. Förster (fluorescence) resonance energy transfer (FRET)-based methods are excellent tools for determining proximity and supramolecular organization of biomolecules at the cell surface or inside the cell. This could well be the basis for the increasing popularity of FRET. In fact, the number of publications exploiting FRET has exploded since the turn of the millennium. Interestingly, most applications are microscope-based, and only a fraction employs flow cytometry, even though the latter offers great statistical power owed to the potentially huge number of individually measured cells. However, with the increased availability of multi-laser flow cytometers, strategies to obtain absolute FRET efficiencies can now be relatively facilely implemented. In this chapter, we intend to provide generally useable protocols for measuring FRET in flow cytometry. After a concise theoretical introduction, recipes are provided for successful labeling techniques and measurement approaches. The simple, quenching-based population-level measurement, the classic ratiometric, intensity-based technique providing cell-by-cell actual FRET efficiencies, and a more advanced version of the latter, allowing for cell-by-cell autofluorescence correction are described. An Excel macro pre-loaded with spectral data of the most commonly used fluorophores is also provided for easy calculation of average FRET efficiencies. Finally, points of caution are given to help design proper experiments and critically interpret the results.
Collapse
Affiliation(s)
- László Ujlaky-Nagy
- Department of Biophysics and Cell Biology, MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
27
|
Lagendijk AK, Gomez GA, Baek S, Hesselson D, Hughes WE, Paterson S, Conway DE, Belting HG, Affolter M, Smith KA, Schwartz MA, Yap AS, Hogan BM. Live imaging molecular changes in junctional tension upon VE-cadherin in zebrafish. Nat Commun 2017; 8:1402. [PMID: 29123087 PMCID: PMC5680264 DOI: 10.1038/s41467-017-01325-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/08/2017] [Indexed: 12/11/2022] Open
Abstract
Forces play diverse roles in vascular development, homeostasis and disease. VE-cadherin at endothelial cell-cell junctions links the contractile acto-myosin cytoskeletons of adjacent cells, serving as a tension-transducer. To explore tensile changes across VE-cadherin in live zebrafish, we tailored an optical biosensor approach, originally established in vitro. We validate localization and function of a VE-cadherin tension sensor (TS) in vivo. Changes in tension across VE-cadherin observed using ratio-metric or lifetime FRET measurements reflect acto-myosin contractility within endothelial cells. Furthermore, we apply the TS to reveal biologically relevant changes in VE-cadherin tension that occur as the dorsal aorta matures and upon genetic and chemical perturbations during embryonic development. Mechanical forces play a crucial role during morphogenesis, but how these are sensed and transduced in vivo is not fully understood. Here the authors apply a FRET tension sensor to live zebrafish and study changes in VE-cadherin tension at endothelial cell-cell junctions during arterial maturation.
Collapse
Affiliation(s)
- Anne Karine Lagendijk
- Institute for Molecular Bioscience, Genomics of Development and Disease division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia.
| | - Guillermo A Gomez
- Institute for Molecular Bioscience, Cell Biology and Molecular Medicine division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia.,Centre for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, 5000, SA, Australia
| | - Sungmin Baek
- Institute for Molecular Bioscience, Genomics of Development and Disease division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia
| | - Daniel Hesselson
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, 2010, NSW, Australia
| | - William E Hughes
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, 2010, NSW, Australia
| | - Scott Paterson
- Institute for Molecular Bioscience, Genomics of Development and Disease division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 70, 4056, Basel, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 70, 4056, Basel, Switzerland
| | - Kelly A Smith
- Institute for Molecular Bioscience, Genomics of Development and Disease division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia
| | - Martin A Schwartz
- Yale Cardiovascular Research Center and Department of Internal Medicine, Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Alpha S Yap
- Institute for Molecular Bioscience, Cell Biology and Molecular Medicine division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, Genomics of Development and Disease division, The University of Queensland, 306 Carmody Road, St Lucia, 4072, QLD, Australia
| |
Collapse
|
28
|
Almada E, Tonucci FM, Hidalgo F, Ferretti A, Ibarra S, Pariani A, Vena R, Favre C, Girardini J, Kierbel A, Larocca MC. Akap350 Recruits Eb1 to The Spindle Poles, Ensuring Proper Spindle Orientation and Lumen Formation in 3d Epithelial Cell Cultures. Sci Rep 2017; 7:14894. [PMID: 29097729 PMCID: PMC5668257 DOI: 10.1038/s41598-017-14241-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/06/2017] [Indexed: 01/08/2023] Open
Abstract
The organization of epithelial cells to form hollow organs with a single lumen requires the accurate three-dimensional arrangement of cell divisions. Mitotic spindle orientation is defined by signaling pathways that provide molecular links between specific spots at the cell cortex and astral microtubules, which have not been fully elucidated. AKAP350 is a centrosomal/Golgi scaffold protein, implicated in the regulation of microtubule dynamics. Using 3D epithelial cell cultures, we found that cells with decreased AKAP350 expression (AKAP350KD) formed polarized cysts with abnormal lumen morphology. Analysis of mitotic cells in AKAP350KD cysts indicated defective spindle alignment. We established that AKAP350 interacts with EB1, a microtubule associated protein that regulates spindle orientation, at the spindle poles. Decrease of AKAP350 expression lead to a significant reduction of EB1 levels at spindle poles and astral microtubules. Conversely, overexpression of EB1 rescued the defective spindle orientation induced by deficient AKAP350 expression. The specific delocalization of the AKAP350/EB1complex from the centrosome decreased EB1 levels at astral microtubules and lead to the formation of 3D-organotypic structures which resembled AKAP350KD cysts. We conclude that AKAP350 recruits EB1 to the spindle poles, ensuring EB1 presence at astral microtubules and proper spindle orientation during epithelial morphogenesis.
Collapse
Affiliation(s)
- Evangelina Almada
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Facundo M Tonucci
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Florencia Hidalgo
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Anabela Ferretti
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Solange Ibarra
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Alejandro Pariani
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Rodrigo Vena
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Cristián Favre
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Javier Girardini
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Arlinet Kierbel
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, CONICET, San Martín, Buenos Aires, Argentina
| | - M Cecilia Larocca
- Instituto de Fisiología Experimental, Consejo de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
29
|
Masukawa D, Koga M, Sezaki A, Nakao Y, Kamikubo Y, Hashimoto T, Okuyama-Oki Y, Aladeokin AC, Nakamura F, Yokoyama U, Wakui H, Ichinose H, Sakurai T, Umemura S, Tamura K, Ishikawa Y, Goshima Y. L-DOPA sensitizes vasomotor tone by modulating the vascular alpha1-adrenergic receptor. JCI Insight 2017; 2:90903. [PMID: 28931752 DOI: 10.1172/jci.insight.90903] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 08/15/2017] [Indexed: 11/17/2022] Open
Abstract
Blood pressure is regulated by extrinsic factors including noradrenaline, the sympathetic neurotransmitter that controls cardiovascular functions through adrenergic receptors. However, the fine-tuning system of noradrenaline signaling is relatively unknown. We here show that l-3,4-dihydroxyphenylalanine (L-DOPA), a precursor of catecholamines, sensitizes the vascular adrenergic receptor alpha1 (ADRA1) through activation of L-DOPA receptor GPR143. In WT mice, intravenous infusion of the ADRA1 agonist phenylephrine induced a transient elevation of blood pressure. This response was attenuated in Gpr143 gene-deficient (Gpr143-/y) mice. Specific knockout of Gpr143 in vascular smooth muscle cells (VSMCs) also showed a similar phenotype, indicating that L-DOPA directly modulates ADRA1 signaling in the VSMCs. L-DOPA at nanomolar concentrations alone produced no effect on the VSMCs, but it enhanced phenylephrine-induced vasoconstriction and intracellular Ca2+ responses. Phenylephrine also augmented the phosphorylation of extracellular signal-regulated kinases in cultured VSMCs from WT but not Gpr143-/y mice. In WT mice, blood pressure increased during the transition from light-rest to dark-active phases. This elevation was not observed in Gpr143-/y mice. Taken together, our findings provide evidence for L-DOPA/GPR143 signaling that exerts precursor control of sympathetic neurotransmission through sensitizing vascular ADRA1.
Collapse
Affiliation(s)
- Daiki Masukawa
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Motokazu Koga
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Anna Sezaki
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuka Nakao
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuji Kamikubo
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tatsuo Hashimoto
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Medical Science and Cardiorenal Medicine, and
| | | | - Aderemi Caleb Aladeokin
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Hiroshi Ichinose
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Takashi Sakurai
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
30
|
Lang MA, Jenkins SA, Balzano P, Owoyele A, Patel A, Bamezai AK. Engaging Ly-6A/Sca-1 triggers lipid raft-dependent and -independent responses in CD4 + T-cell lines. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:448-460. [PMID: 28660664 PMCID: PMC5691314 DOI: 10.1002/iid3.182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The lymphocyte antigen 6 (Ly-6) supergene family encodes proteins of 12-14 kda in molecular mass that are either secreted or anchored to the plasma membrane through a glycosyl-phosphatidylinisotol (GPI) lipid anchor at the carboxy-terminus. The lipidated GPI-anchor allows localization of Ly-6 proteins to the 10-100 nm cholesterol-rich nano-domains on the membrane, also known as lipid rafts. Ly-6A/Sca-1, a member of Ly-6 gene family is known to transduce signals despite the absence of transmembrane and cytoplasmic domains. It is hypothesized that the localization of Ly-6A/Sca-1 with in lipid rafts allows this protein to transduce signals to the cell interior. METHODS AND RESULTS In this study, we found that cross-linking mouse Ly-6A/Sca-1 protein with a monoclonal antibody results in functionally distinct responses that occur simultaneously. Ly-6A/Sca-1 triggered a cell stimulatory response as gauged by cytokine production with a concurrent inhibitory response as indicated by growth inhibition and apoptosis. While production of interleukin 2 (IL-2) cytokine by CD4+ T cell line in response to cross-linking Ly-6A/Sca-1 was dependent on the integrity of lipid rafts, the observed cell death occurred independently of it. Growth inhibited CD4+ T cells showed up-regulated expression of the inhibitory cell cycle protein p27kip but not of p53. In addition, Ly-6A/Sca-1 induced translocation of cytochrome C to the cytoplasm along with activated caspase 3 and caspase 9, thereby suggesting an intrinsic apoptotic cell death mechanism. CONCLUSIONS We conclude that opposing responses with differential dependence on the integrity of lipid rafts are triggered by engaging Ly-6A/Sca-1 protein on the membrane of transformed CD4+ T cells.
Collapse
Affiliation(s)
- Melissa A Lang
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Sultan A Jenkins
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Phillip Balzano
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Adeyinka Owoyele
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Akshay Patel
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Anil K Bamezai
- Department of Biology, Villanova University, Villanova, Pennsylvania
| |
Collapse
|
31
|
Stone MB, Shelby SA, Núñez MF, Wisser K, Veatch SL. Protein sorting by lipid phase-like domains supports emergent signaling function in B lymphocyte plasma membranes. eLife 2017; 6. [PMID: 28145867 PMCID: PMC5373823 DOI: 10.7554/elife.19891] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/31/2017] [Indexed: 12/19/2022] Open
Abstract
Diverse cellular signaling events, including B cell receptor (BCR) activation, are hypothesized to be facilitated by domains enriched in specific plasma membrane lipids and proteins that resemble liquid-ordered phase-separated domains in model membranes. This concept remains controversial and lacks direct experimental support in intact cells. Here, we visualize ordered and disordered domains in mouse B lymphoma cell membranes using super-resolution fluorescence localization microscopy, demonstrate that clustered BCR resides within ordered phase-like domains capable of sorting key regulators of BCR activation, and present a minimal, predictive model where clustering receptors leads to their collective activation by stabilizing an extended ordered domain. These results provide evidence for the role of membrane domains in BCR signaling and a plausible mechanism of BCR activation via receptor clustering that could be generalized to other signaling pathways. Overall, these studies demonstrate that lipid mediated forces can bias biochemical networks in ways that broadly impact signal transduction. DOI:http://dx.doi.org/10.7554/eLife.19891.001 Membranes made of molecules called lipids surround every living cell to protect the cell's contents. Cells also communicate with the outside environment via their membranes. Proteins in the membrane receive information from the environment and trigger signaling pathways inside the cell to relay this information to the center of cell. The way in which proteins are organized on the membrane has a major influence on their signaling activity. Some areas of the membrane are more crowded with certain lipids and signaling proteins than others. Lipid and protein molecules of particular types can come together and form distinct areas called “ordered” and “disordered” domains. The lipids in ordered domains are more tightly packed than disordered domains and it is thought that this difference allows domains to selectively exclude or include certain proteins. Ordered domains are also known as "lipid rafts". Lipid rafts and disordered domains may help cells to control the activities of signaling pathways, however, technical limitations have made it difficult to study the roles of these domains. The membranes surrounding immune cells called B cells contain a protein called the B cell receptor, which engages with proteins from microbes and other foreign invaders. When the B cell receptor binds to a foreign protein it forms clusters with other B cell receptors and becomes active, triggering a signaling pathway that leads to immune responses. Stone, Shelby et al. examined lipid rafts and disordered domains in B cells from mice using a technique called super-resolution fluorescence microscopy. The results show that clusters of B cell receptors are present within lipid rafts. These clusters made the lipid rafts larger and more stable. A protein that is needed during the early stages of B cell receptor signaling was also found in the same lipid rafts. Another protein that terminates signaling was excluded because it prefers disordered domains. Together, this provides a local environment in certain areas of the membrane that favors receptor activity and supports the subsequent immune response. Future work is needed to understand how cells control the make-up of lipids and proteins within their membranes, and how defects in this regulation can alter signaling activity and lead to disease. DOI:http://dx.doi.org/10.7554/eLife.19891.002
Collapse
Affiliation(s)
- Matthew B Stone
- Department of Biophysics, University of Michigan, Ann Arbor, United States
| | - Sarah A Shelby
- Department of Biophysics, University of Michigan, Ann Arbor, United States
| | - Marcos F Núñez
- Department of Biophysics, University of Michigan, Ann Arbor, United States
| | - Kathleen Wisser
- Department of Biophysics, University of Michigan, Ann Arbor, United States
| | - Sarah L Veatch
- Department of Biophysics, University of Michigan, Ann Arbor, United States
| |
Collapse
|
32
|
Triantafilou M, Triantafilou K. Receptor cluster formation during activation by bacterial products. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519030090051001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The recognition of bacterial products, such as lipopolysaccharide (LPS) by the innate immune system lead to a strong pro-inflammatory response that can eventually lead to fatal sepsis syndrome in humans. Although CD14 and TLR4 have been identified as the key molecules involved in LPSinduced signal transduction, accumulating evidence indicates that multiple receptors are also involved. Our group has recently identified a cluster of receptors, involving heat-shock proteins 70 and 90, chemokine receptor 4 as well as growth differentiation factor 5, that are formed following LPS stimulation. In addition, we present data demonstrating that these molecules associate with TLR4 and accumulate in membrane microdomains following LPS ligation. Our results suggest that the entire bacterial recognition is based around the recruitment of multiple signalling molecules, in addition to CD14 and TLRs, within the lipid rafts. We propose that different combinational associations of receptors within activation clusters determine the different responses to a variety of bacterial stimuli.
Collapse
Affiliation(s)
- Martha Triantafilou
- Institute of Biomedical and Biomolecular Sciences, School of Biological Sciences, University of Portsmouth, Portsmouth, UK
| | - Kathy Triantafilou
- Institute of Biomedical and Biomolecular Sciences, School of Biological Sciences, University of Portsmouth, Portsmouth, UK, arch
| |
Collapse
|
33
|
The multilayer nanoparticles for deep penetration of docetaxel into tumor parenchyma to overcome tumor microenvironment. Colloids Surf B Biointerfaces 2016; 146:833-40. [PMID: 27451372 DOI: 10.1016/j.colsurfb.2016.07.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/13/2016] [Accepted: 07/15/2016] [Indexed: 12/25/2022]
Abstract
Deep penetration of the anticancer drug, docetaxel (DTX), into tumor parenchyma was demonstrated to achieve improved chemotherapy. For this purpose, a multistage nanostructure was designed and characterized using the multilayer nanoparticles (NPs). The multilayer NPs had a core/shell structure. The core was composed of the DTX-loaded Pluronic NPs (diameter: 12nm) that were transferred into the inner side of vesicles to form the vesicle NPs. Förster resonance energy transfer (FRET) in the NPs was observed to verify the incorporation of the DTX-loaded Pluronic NPs into the inner side of the vesicles during the formation of the vesicle NPs. Subsequently, the vesicle NPs were stabilized through Pluronic-lipid bilayer interaction to form the multilayer NPs. To examine the morphology and size distribution of the multilayer NPs, transmittance electron microscopy and dynamic light scattering were used. In vitro release behavior and toxicity were observed to verify the functionality of the multilayer NPs as nanocarriers for cancer therapy. Multistage functionality was evaluated by cellular uptake and tissue distribution behaviors of the multilayer NPs. The biodistribution of the multilayer NPs and their antitumor efficacy were also observed to understand the role of multistage functionality for improved chemotherapy.
Collapse
|
34
|
Hare JE, Goodchild SC, Breit SN, Curmi PMG, Brown LJ. Interaction of Human Chloride Intracellular Channel Protein 1 (CLIC1) with Lipid Bilayers: A Fluorescence Study. Biochemistry 2016; 55:3825-33. [PMID: 27299171 DOI: 10.1021/acs.biochem.6b00080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chloride intracellular channel protein 1 (CLIC1) is very unusual as it adopts a soluble glutathione S-transferase-like canonical fold but can also autoinsert into lipid bilayers to form an ion channel. The conversion between these forms involves a large, but reversible, structural rearrangement of the CLIC1 module. The only identified environmental triggers controlling the metamorphic transition of CLIC1 are pH and oxidation. Until now, there have been no high-resolution structural data available for the CLIC1 integral membrane state, and consequently, a limited understanding of how CLIC1 unfolds and refolds across the bilayer to form a membrane protein with ion channel activity exists. Here we show that fluorescence spectroscopy can be used to establish the interaction and position of CLIC1 in a lipid bilayer. Our method employs a fluorescence energy transfer (FRET) approach between CLIC1 and a dansyl-labeled lipid analogue to probe the CLIC1-lipid interface. Under oxidizing conditions, a strong FRET signal between the single tryptophan residue of CLIC1 (Trp35) and the dansyl-lipid analogue was detected. When considering the proportion of CLIC1 interacting with the lipid bilayer, as estimated by fluorescence quenching experiments, the FRET distance between Trp35 and the dansyl moiety on the membrane surface was determined to be ∼15 Å. This FRET-detected interaction provides direct structural evidence that CLIC1 associates with membranes. The results presented support the current model of an oxidation-driven interaction of CLIC1 with lipid bilayers and also propose a membrane anchoring role for Trp35.
Collapse
Affiliation(s)
- Joanna E Hare
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Sophia C Goodchild
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds , Leeds LS29JT, United Kingdom
| | - Samuel N Breit
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital , Sydney, New South Wales 2010, Australia
| | - Paul M G Curmi
- School of Physics, University of New South Wales , Sydney, New South Wales 2052, Australia
| | - Louise J Brown
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| |
Collapse
|
35
|
Zhou M, Li Q, Wang R. Current Experimental Methods for Characterizing Protein-Protein Interactions. ChemMedChem 2016; 11:738-56. [PMID: 26864455 PMCID: PMC7162211 DOI: 10.1002/cmdc.201500495] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/08/2016] [Indexed: 12/14/2022]
Abstract
Protein molecules often interact with other partner protein molecules in order to execute their vital functions in living organisms. Characterization of protein-protein interactions thus plays a central role in understanding the molecular mechanism of relevant protein molecules, elucidating the cellular processes and pathways relevant to health or disease for drug discovery, and charting large-scale interaction networks in systems biology research. A whole spectrum of methods, based on biophysical, biochemical, or genetic principles, have been developed to detect the time, space, and functional relevance of protein-protein interactions at various degrees of affinity and specificity. This article presents an overview of these experimental methods, outlining the principles, strengths and limitations, and recent developments of each type of method.
Collapse
Affiliation(s)
- Mi Zhou
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China
| | - Qing Li
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China
| | - Renxiao Wang
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macau, 999078, People's Republic of China.
| |
Collapse
|
36
|
Zhou M, Li Q, Wang R. Current Experimental Methods for Characterizing Protein-Protein Interactions. ChemMedChem 2016. [PMID: 26864455 DOI: 10.1002/cmdc.201500495.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Protein molecules often interact with other partner protein molecules in order to execute their vital functions in living organisms. Characterization of protein-protein interactions thus plays a central role in understanding the molecular mechanism of relevant protein molecules, elucidating the cellular processes and pathways relevant to health or disease for drug discovery, and charting large-scale interaction networks in systems biology research. A whole spectrum of methods, based on biophysical, biochemical, or genetic principles, have been developed to detect the time, space, and functional relevance of protein-protein interactions at various degrees of affinity and specificity. This article presents an overview of these experimental methods, outlining the principles, strengths and limitations, and recent developments of each type of method.
Collapse
Affiliation(s)
- Mi Zhou
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China
| | - Qing Li
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China
| | - Renxiao Wang
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd, Shanghai, 200032, People's Republic of China. .,State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macau, 999078, People's Republic of China.
| |
Collapse
|
37
|
Tran J, Magenau A, Rodriguez M, Rentero C, Royo T, Enrich C, Thomas SR, Grewal T, Gaus K. Activation of Endothelial Nitric Oxide (eNOS) Occurs through Different Membrane Domains in Endothelial Cells. PLoS One 2016; 11:e0151556. [PMID: 26977592 PMCID: PMC4792450 DOI: 10.1371/journal.pone.0151556] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 03/01/2016] [Indexed: 11/18/2022] Open
Abstract
Endothelial cells respond to a large range of stimuli including circulating lipoproteins, growth factors and changes in haemodynamic mechanical forces to regulate the activity of endothelial nitric oxide synthase (eNOS) and maintain blood pressure. While many signalling pathways have been mapped, the identities of membrane domains through which these signals are transmitted are less well characterized. Here, we manipulated bovine aortic endothelial cells (BAEC) with cholesterol and the oxysterol 7-ketocholesterol (7KC). Using a range of microscopy techniques including confocal, 2-photon, super-resolution and electron microscopy, we found that sterol enrichment had differential effects on eNOS and caveolin-1 (Cav1) colocalisation, membrane order of the plasma membrane, caveolae numbers and Cav1 clustering. We found a correlation between cholesterol-induced condensation of the plasma membrane and enhanced high density lipoprotein (HDL)-induced eNOS activity and phosphorylation suggesting that cholesterol domains, but not individual caveolae, mediate HDL stimulation of eNOS. Vascular endothelial growth factor (VEGF)-induced and shear stress-induced eNOS activity was relatively independent of membrane order and may be predominantly controlled by the number of caveolae on the cell surface. Taken together, our data suggest that signals that activate and phosphorylate eNOS are transmitted through distinct membrane domains in endothelial cells.
Collapse
Affiliation(s)
- Jason Tran
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Astrid Magenau
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Macarena Rodriguez
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
| | - Carles Rentero
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Teresa Royo
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Shane R. Thomas
- School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Thomas Grewal
- Faculty of Pharmacy A15, University of Sydney, Sydney, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
38
|
Pezzarossa A, Zosel F, Schmidt T. Visualization of HRas Domains in the Plasma Membrane of Fibroblasts. Biophys J 2016; 108:1870-7. [PMID: 25902427 DOI: 10.1016/j.bpj.2015.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 01/07/2015] [Accepted: 03/04/2015] [Indexed: 11/19/2022] Open
Abstract
The plasma membrane is a highly complex, organized structure where the lateral organization of signaling proteins is tightly regulated. In the case of Ras proteins, it has been suggested that the differential activity of the various isoforms is due to protein localization in separate membrane compartments. To date, direct visualization of such compartmentalization has been achieved only by electron microscopy on membrane sheets. Here, we combine photoactivated light microscopy with quantitative statistical analysis to visualize protein distribution in intact cells. In particular, we focus on the localization of HRas and its minimal anchoring domain, CAAX. We demonstrate the existence of a complex partitioning behavior, where small domains coexist with larger ones. The protein content in these domains varied from two molecules to tens of molecules. We found that 40% of CAAX and 60% of HRas were localized in domains. Subsequently, we were able to manipulate protein distributions by inducing coalescence of supposedly cholesterol-enriched domains. Clustering resulted in an increase of the localized fraction by 15%.
Collapse
Affiliation(s)
- Anna Pezzarossa
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, The Netherlands
| | - Franziska Zosel
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, The Netherlands
| | - Thomas Schmidt
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, The Netherlands.
| |
Collapse
|
39
|
Barroso M, Tucker H, Drake L, Nichol K, Drake JR. Antigen-B Cell Receptor Complexes Associate with Intracellular major histocompatibility complex (MHC) Class II Molecules. J Biol Chem 2015; 290:27101-27112. [PMID: 26400081 DOI: 10.1074/jbc.m115.649582] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Indexed: 01/09/2023] Open
Abstract
Antigen processing and MHC class II-restricted antigen presentation by antigen-presenting cells such as dendritic cells and B cells allows the activation of naïve CD4+ T cells and cognate interactions between B cells and effector CD4+ T cells, respectively. B cells are unique among class II-restricted antigen-presenting cells in that they have a clonally restricted antigen-specific receptor, the B cell receptor (BCR), which allows the cell to recognize and respond to trace amounts of foreign antigen present in a sea of self-antigens. Moreover, engagement of peptide-class II complexes formed via BCR-mediated processing of cognate antigen has been shown to result in a unique pattern of B cell activation. Using a combined biochemical and imaging/FRET approach, we establish that internalized antigen-BCR complexes associate with intracellular class II molecules. We demonstrate that the M1-paired MHC class II conformer, shown previously to be critical for CD4 T cell activation, is incorporated selectively into these complexes and loaded selectively with peptide derived from BCR-internalized cognate antigen. These results demonstrate that, in B cells, internalized antigen-BCR complexes associate with intracellular MHC class II molecules, potentially defining a site of class II peptide acquisition, and reveal a selective role for the M1-paired class II conformer in the presentation of cognate antigen. These findings provide key insights into the molecular mechanisms used by B cells to control the source of peptides charged onto class II molecules, allowing the immune system to mount an antibody response focused on BCR-reactive cognate antigen.
Collapse
Affiliation(s)
- Margarida Barroso
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York 12208
| | - Heidi Tucker
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Lisa Drake
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - Kathleen Nichol
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208
| | - James R Drake
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York 12208.
| |
Collapse
|
40
|
Saha S, Anilkumar AA, Mayor S. GPI-anchored protein organization and dynamics at the cell surface. J Lipid Res 2015; 57:159-75. [PMID: 26394904 DOI: 10.1194/jlr.r062885] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Indexed: 01/05/2023] Open
Abstract
The surface of eukaryotic cells is a multi-component fluid bilayer in which glycosylphosphatidylinositol (GPI)-anchored proteins are an abundant constituent. In this review, we discuss the complex nature of the organization and dynamics of GPI-anchored proteins at multiple spatial and temporal scales. Different biophysical techniques have been utilized for understanding this organization, including fluorescence correlation spectroscopy, fluorescence recovery after photobleaching, single particle tracking, and a number of super resolution methods. Major insights into the organization and dynamics have also come from exploring the short-range interactions of GPI-anchored proteins by fluorescence (or Förster) resonance energy transfer microscopy. Based on the nanometer to micron scale organization, at the microsecond to the second time scale dynamics, a picture of the membrane bilayer emerges where the lipid bilayer appears inextricably intertwined with the underlying dynamic cytoskeleton. These observations have prompted a revision of the current models of plasma membrane organization, and suggest an active actin-membrane composite.
Collapse
Affiliation(s)
- Suvrajit Saha
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore 560065, India
| | - Anupama Ambika Anilkumar
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore 560065, India Shanmugha Arts, Science, Technology and Research Academy, Thanjavur 613401, India
| | - Satyajit Mayor
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore 560065, India Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore 560065, India
| |
Collapse
|
41
|
Lateral diffusion contributes to FRET from lanthanide-tagged membrane proteins. Biochem Biophys Res Commun 2015; 464:244-8. [PMID: 26119691 DOI: 10.1016/j.bbrc.2015.06.127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 11/20/2022]
Abstract
Diffusion can enhance Förster resonance energy transfer (FRET) when donors or acceptors diffuse distances that are similar to the distances separating them during the donor's excited state lifetime. Lanthanide donors remain in the excited state for milliseconds, which makes them useful for time-resolved FRET applications but also allows time for diffusion to enhance energy transfer. Here we show that diffusion dramatically enhances FRET between membrane proteins labeled with lanthanide donors. This phenomenon complicates interpretation of experiments that use long-lived donors to infer association or proximity of mobile membrane proteins, but also offers a method of monitoring diffusion in membrane domains in real time in living cells.
Collapse
|
42
|
Complex formation and functional interaction between adenosine A1 receptor and type-1 metabotropic glutamate receptor. J Pharmacol Sci 2015; 128:125-30. [PMID: 26154847 DOI: 10.1016/j.jphs.2015.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/24/2022] Open
Abstract
The adenosine A1 receptor (A1R) is a G protein-coupled receptor (GPCR) for adenosine, a ubiquitous neuromodulator, and thus regulates neuronal excitability, as well as arousal and sensitivity to pain. In addition, we have previously described a new mode of action for A1R: in cerebellar Purkinje cells, its activation attenuates neuronal responses to glutamate, as mediated by the type-1 metabotropic glutamate receptor (mGluR1). mGluR1 is also a GPCR, and elicits such responses as long-term depression of the postsynaptic response to glutamate, a cellular basis for cerebellar motor learning. Here, we explore in greater detail the interaction between A1R and mGluR1 using non-neuronal cells. Co-immunoprecipitation and Förster resonance energy transfer (FRET) analysis reveal that A1R and mGluR1 form a complex. Furthermore, we found that mGluR1 activation inhibits A1R signaling, as measured by changes in intracellular cAMP. These findings demonstrate that A1R and mGluR1 have the intrinsic ability to form a heteromeric complex and mutually modulate signaling. This interaction may represent a new form of intriguing GPCR-mediated cellular responses.
Collapse
|
43
|
BRET evidence that β2 adrenergic receptors do not oligomerize in cells. Sci Rep 2015; 5:10166. [PMID: 25955971 PMCID: PMC4424835 DOI: 10.1038/srep10166] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/01/2015] [Indexed: 11/29/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is often used to study association of membrane proteins, and in particular oligomerization of G protein-coupled receptors (GPCRs). Oligomerization of class A GPCRs is controversial, in part because the methods used to study this question are not completely understood. Here we reconsider oligomerization of the class A β2 adrenergic receptor (β2AR), and reevaluate BRET titration as a method to study membrane protein association. Using inducible expression of the energy acceptor at multiple levels of donor expression we find that BRET between β2AR protomers is directly proportional to the density of the acceptor up to ~3,000 acceptors μm−2, and does not depend on the density of the donor or on the acceptor:donor (A:D) stoichiometry. In contrast, BRET between tightly-associating control proteins does not depend on the density of the acceptor, but does depend on the density of the donor and on the A:D ratio. We also find that the standard frameworks used to interpret BRET titration experiments rely on simplifying assumptions that are frequently invalid. These results suggest that β2ARs do not oligomerize in cells, and demonstrate a reliable method of assessing membrane protein association with BRET.
Collapse
|
44
|
Sevcsik E, Brameshuber M, Fölser M, Weghuber J, Honigmann A, Schütz GJ. GPI-anchored proteins do not reside in ordered domains in the live cell plasma membrane. Nat Commun 2015; 6:6969. [PMID: 25897971 PMCID: PMC4430820 DOI: 10.1038/ncomms7969] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 03/18/2015] [Indexed: 02/07/2023] Open
Abstract
The organization of proteins and lipids in the plasma membrane has been the subject of a long-lasting debate. Membrane rafts of higher lipid chain order were proposed to mediate protein interactions, but have thus far not been directly observed. Here we use protein micropatterning combined with single-molecule tracking to put current models to the test: we rearranged lipid-anchored raft proteins (glycosylphosphatidylinositol(GPI)-anchored-mGFP) directly in the live cell plasma membrane and measured the effect on the local membrane environment. Intriguingly, this treatment does neither nucleate the formation of an ordered membrane phase nor result in any enrichment of nanoscopic-ordered domains within the micropatterned regions. In contrast, we find that immobilized mGFP-GPIs behave as inert obstacles to the diffusion of other membrane constituents without influencing their membrane environment over distances beyond their physical size. Our results indicate that phase partitioning is not a fundamental element of protein organization in the plasma membrane.
Collapse
Affiliation(s)
- Eva Sevcsik
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| | - Mario Brameshuber
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| | - Martin Fölser
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| | - Julian Weghuber
- School of Engineering and Environmental Sciences, University of Applied Sciences Upper Austria, Stelzhamerstrasse 23, Wels 4600, Austria
| | - Alf Honigmann
- Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry, Am Fassberg 11, Göttingen 37077, Germany
| | - Gerhard J Schütz
- Institute of Applied Physics, Vienna University of Technology, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| |
Collapse
|
45
|
Curthoys NM, Parent M, Mlodzianoski M, Nelson AJ, Lilieholm J, Butler MB, Valles M, Hess ST. Dances with Membranes: Breakthroughs from Super-resolution Imaging. CURRENT TOPICS IN MEMBRANES 2015; 75:59-123. [PMID: 26015281 PMCID: PMC5584789 DOI: 10.1016/bs.ctm.2015.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biological membrane organization mediates numerous cellular functions and has also been connected with an immense number of human diseases. However, until recently, experimental methodologies have been unable to directly visualize the nanoscale details of biological membranes, particularly in intact living cells. Numerous models explaining membrane organization have been proposed, but testing those models has required indirect methods; the desire to directly image proteins and lipids in living cell membranes is a strong motivation for the advancement of technology. The development of super-resolution microscopy has provided powerful tools for quantification of membrane organization at the level of individual proteins and lipids, and many of these tools are compatible with living cells. Previously inaccessible questions are now being addressed, and the field of membrane biology is developing rapidly. This chapter discusses how the development of super-resolution microscopy has led to fundamental advances in the field of biological membrane organization. We summarize the history and some models explaining how proteins are organized in cell membranes, and give an overview of various super-resolution techniques and methods of quantifying super-resolution data. We discuss the application of super-resolution techniques to membrane biology in general, and also with specific reference to the fields of actin and actin-binding proteins, virus infection, mitochondria, immune cell biology, and phosphoinositide signaling. Finally, we present our hopes and expectations for the future of super-resolution microscopy in the field of membrane biology.
Collapse
Affiliation(s)
- Nikki M. Curthoys
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Matthew Parent
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | | | - Andrew J. Nelson
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Jennifer Lilieholm
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Michael B. Butler
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Matthew Valles
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Samuel T. Hess
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| |
Collapse
|
46
|
Paladino S, Lebreton S, Zurzolo C. Trafficking and Membrane Organization of GPI-Anchored Proteins in Health and Diseases. CURRENT TOPICS IN MEMBRANES 2015; 75:269-303. [PMID: 26015286 DOI: 10.1016/bs.ctm.2015.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are a class of lipid-anchored proteins attached to the membranes by a glycolipid anchor that is added, as posttranslation modification, in the endoplasmic reticulum. GPI-APs are expressed at the cell surface of eukaryotes where they play diverse vital functions. Like all plasma membrane proteins, GPI-APs must be correctly sorted along the different steps of the secretory pathway to their final destination. The presence of both a glycolipid anchor and a protein portion confers special trafficking features to GPI-APs. Here, we discuss the recent advances in the field of GPI-AP trafficking, focusing on the mechanisms regulating their biosynthetic pathway and plasma membrane organization. We also discuss how alterations of these mechanisms can result in different diseases. Finally, we will examine the strict relationship between the trafficking and function of GPI-APs in epithelial cells.
Collapse
Affiliation(s)
- Simona Paladino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Napoli, Italy; CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Stéphanie Lebreton
- Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur, Paris, France
| | - Chiara Zurzolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Napoli, Italy; Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur, Paris, France
| |
Collapse
|
47
|
Mazurkiewicz JE, Herrick-Davis K, Barroso M, Ulloa-Aguirre A, Lindau-Shepard B, Thomas RM, Dias JA. Single-molecule analyses of fully functional fluorescent protein-tagged follitropin receptor reveal homodimerization and specific heterodimerization with lutropin receptor. Biol Reprod 2015; 92:100. [PMID: 25761594 DOI: 10.1095/biolreprod.114.125781] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/02/2015] [Indexed: 01/27/2023] Open
Abstract
We have previously shown that the carboxyl terminus (cT) of human follicle-stimulating hormone (FSH, follitropin) receptor (FSHR) is clipped before insertion into the plasma membrane. Surprisingly, several different constructs of FSHR fluorescent fusion proteins (FSHR-FPs) failed to traffic to the plasma membrane. Subsequently, we discovered that substituting the extreme cT of luteinizing hormone (LH) receptor (LHR) to create an FSHR-LHRcT chimera has no effect on FSHR functionality. Therefore, we used this approach to create an FSHR-LHRcT-FP fusion. We found this chimeric FSHR-LHRcT-FP was expressed in HEK293 cells at levels similar to reported values for FSHR in human granulosa cells, bound FSH with high affinity, and transduced FSH binding to produce cAMP. Quantitative fluorescence resonance energy transfer (FRET) analysis of FSHR-LHRcT-YFP/FSHR-LHRcT-mCherry pairs revealed an average FRET efficiency of 12.9 ± 5.7. Advanced methods in single-molecule analyses were applied in order to ascertain the oligomerization state of the FSHR-LHRcT. Fluorescence correlation spectroscopy coupled with photon-counting histogram analyses demonstrated that the FSHR-LHRcT-FP fusion protein exists as a freely diffusing homodimer in the plasma membrane. A central question is whether LHR could oligomerize with FSHR, because both receptors are coexpressed in differentiated granulosa cells. Indeed, FRET analysis revealed an average FRET efficiency of 14.4 ± 7.5 when the FSHR-LHR cT-mCherry was coexpressed with LHR-YFP. In contrast, coexpression of a 5-HT2cVSV-YFP with FSHR-LHR cT-mCherry showed only 5.6 ± 3.2 average FRET efficiency, a value indistinguishable from the detection limit using intensity-based FRET methods. These data demonstrate that coexpression of FSHR and LHR can lead to heterodimerization, and we hypothesize that it is possible for this to occur during granulosa cell differentiation.
Collapse
Affiliation(s)
- Joseph E Mazurkiewicz
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York
| | | | - Margarida Barroso
- Center for Cardiovascular Science, Albany Medical College, Albany, New York
| | - Alfredo Ulloa-Aguirre
- Research Support Network, Instituto Nacional de Ciencias Médicas y Nutrición SZ-Universidad Nacional Autónoma de México, México D.F., México
| | - Barbara Lindau-Shepard
- Division of Genetic Disorders, Wadsworth Center, New York State Department of Health, Albany, New York
| | - Richard M Thomas
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York
| | - James A Dias
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York
| |
Collapse
|
48
|
Edidin M. Light and life in Baltimore--and beyond. Biophys J 2015; 108:466-70. [PMID: 25650914 DOI: 10.1016/j.bpj.2014.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 12/07/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022] Open
Abstract
Baltimore has been the home of numerous biophysical studies using light to probe cells. One such study, quantitative measurement of lateral diffusion of rhodopsin, set the standard for experiments in which recovery after photobleaching is used to measure lateral diffusion. Development of this method from specialized microscopes to commercial scanning confocal microscopes has led to widespread use of the technique to measure lateral diffusion of membrane proteins and lipids, and as well diffusion and binding interactions in cell organelles and cytoplasm. Perturbation of equilibrium distributions by photobleaching has also been developed into a robust method to image molecular proximity in terms of fluorescence resonance energy transfer between donor and acceptor fluorophores.
Collapse
Affiliation(s)
- Michael Edidin
- Biology Department, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
49
|
Actin-myosin network is required for proper assembly of influenza virus particles. Virology 2015; 476:141-150. [DOI: 10.1016/j.virol.2014.12.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/02/2014] [Accepted: 12/08/2014] [Indexed: 01/06/2023]
|
50
|
Schieffer D, Naware S, Bakun W, Bamezai AK. Lipid raft-based membrane order is important for antigen-specific clonal expansion of CD4(+) T lymphocytes. BMC Immunol 2014; 15:58. [PMID: 25494999 PMCID: PMC4270042 DOI: 10.1186/s12865-014-0058-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 11/24/2014] [Indexed: 11/24/2022] Open
Abstract
Background Lipid rafts are cholesterol and saturated lipid-rich, nanometer sized membrane domains that are hypothesized to play an important role in compartmentalization and spatiotemporal regulation of cellular signaling. Lipid rafts contribute to the plasma membrane order and to its spatial asymmetry, as well. The raft nanodomains on the surface of CD4+ T lymphocytes coalesce during their interaction with antigen presenting cells (APCs). Sensing of foreign antigen by the antigen receptor on CD4+ T cells occurs during these cell-cell interactions. In response to foreign antigen the CD4+ T cells proliferate, allowing the expansion of few antigen-specific primary CD4+ T cell clones. Proliferating CD4+ T cells specialize in their function by undergoing differentiation into appropriate effectors tailored to mount an effective adaptive immune response against the invading pathogen. Results To investigate the role of lipid raft-based membrane order in the clonal expansion phase of primary CD4+ T cells, we have disrupted membrane order by incorporating an oxysterol, 7-ketocholesterol (7-KC), into the plasma membrane of primary CD4+ T cells expressing a T cell receptor specific to chicken ovalbumin323–339 peptide sequence and tested their antigen-specific response. We report that 7-KC, at concentrations that disrupt lipid rafts, significantly diminish the c-Ovalbumin323–339 peptide-specific clonal expansion of primary CD4+ T cells. Conclusions Our findings suggest that lipid raft-based membrane order is important for clonal expansion of CD4+ T cells in response to a model peptide. Electronic supplementary material The online version of this article (doi:10.1186/s12865-014-0058-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Schieffer
- Department of Biology, Villanova University, 800 Lancaster Avenue, Villanova, PA, 19085, USA. .,Current Address: DeNovix Inc, Wilmington, DE, 19808, USA.
| | - Sanya Naware
- Department of Biology, Villanova University, 800 Lancaster Avenue, Villanova, PA, 19085, USA. .,Current Address: M.D. Program, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA.
| | - Walter Bakun
- Department of Biology, Villanova University, 800 Lancaster Avenue, Villanova, PA, 19085, USA.
| | - Anil K Bamezai
- Department of Biology, Villanova University, 800 Lancaster Avenue, Villanova, PA, 19085, USA.
| |
Collapse
|