1
|
Qiao P, Zhao M, Zhao J, Wen J, Zhao C, Zhang M. Unveiling the camelina MBOAT gene family: Phylogenetic insights and regulatory landscape. Gene 2025; 936:149085. [PMID: 39542282 DOI: 10.1016/j.gene.2024.149085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
The membrane-bound O-acyltransferase (MBOAT) gene family comprises enzymes responsible for transferring acyl groups to various lipid molecules. Some members of the MBOAT gene family and their functions have been extensively studied in the model plant Arabidopsis. However, research on the MBOAT gene family in camelina is still limited. In this study, 54 MBOATs were identified on 17 chromosomes and one unidentified scaffold in camelina, including seven newly identified genes. A total of 149 MBOATs were identified in 10 other species. Six subgroups of these MBOATs with different conservation were classified by phylogenetic analysis, showing diversification between monocots and dicots. Detailed analysis of the motif composition, evolutionary relationships, and gene structures of CsaMBOATs are presented. The results of the syntenic analysis suggest that the evolution of CsaMBOAT gene family is primarily driven by segmental and tandem duplications, and that there is a stronger collinearity within dicots. In addition, analysis of CsaMBOAT gene promoter cis-elements reveals a possible transcriptional regulation and tissue-specific expression, highlighting potential role in plant stress responses and hormone signaling. Furthermore, both the transcriptome and RT-qPCR data revealed the changes in the expression levels of DGAT1 during salt stress treatment. Subsequent analyses indicated that DGAT1 influenced the ratio of fatty acid fractions in the plants. Importantly, a large number of transcription factors involved in the regulation of CsaMBOAT gene expression were identified by WGCNA analysis, and the transcriptional data confirmed that the NAC032 and CAMMTA6 genes play a role upstream of DGAT1. This study not only identified the members of the MBOAT in camelina, but also provided insights and clues into its regulatory mechanisms.
Collapse
Affiliation(s)
- Pengfang Qiao
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Maoqiu Zhao
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jialiang Zhao
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiayin Wen
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Cuizhu Zhao
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meng Zhang
- College of Agronomy, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
2
|
Dharan R, Barnoy A, Tsaturyan AK, Grossman A, Goren S, Yosibash I, Nachmias D, Elia N, Sorkin R, Kozlov MM. Intracellular pressure controls the propagation of tension in crumpled cell membranes. Nat Commun 2025; 16:91. [PMID: 39747015 PMCID: PMC11696741 DOI: 10.1038/s41467-024-55398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Propagation of membrane tension mediates mechanical signal transduction along surfaces of live cells and sets the time scale of mechanical equilibration of cell membranes. Recent studies in several cell types and under different conditions revealed a strikingly wide variation range of the tension propagation speeds including extremely low ones. The latter suggests a possibility of long-living inhomogeneities of membrane tension crucially affecting mechano-sensitive membrane processes. Here, we propose, analyze theoretically, and support experimentally a mechanism of tension propagation in membranes crumpled by the contractile cortical cytoskeleton. The tension spreading is mediated by the membrane flow between the crumples. We predict the pace of the tension propagation to be controlled by the intra-cellular pressure and the degree of the membrane crumpling. We provide experimental support for the suggested mechanism by monitoring the rate of tension propagation in cells exposed to external media of different osmolarities.
Collapse
Affiliation(s)
- Raviv Dharan
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Avishai Barnoy
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Andrey K Tsaturyan
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Alon Grossman
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Shahar Goren
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Yosibash
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Nachmias
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Natalie Elia
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Raya Sorkin
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel.
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.
| | - Michael M Kozlov
- Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv, Israel.
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
3
|
Wang S, Jiang H, Hu M, Gong Y, Zhou H. Evolutionary conservation analysis of human sphingomyelin metabolism pathway genes. Heliyon 2024; 10:e40810. [PMID: 39698091 PMCID: PMC11652929 DOI: 10.1016/j.heliyon.2024.e40810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Sphingomyelin is an important member of the sphingolipid family and was first reported more than a century ago. It has been demonstrated that sphingomyelin plays a crucial role in compositing cell membranes and signaling pathways. Despite extensive functional studies on the sphingolipid metabolism pathway genes, one intriguing question remains: how does the emergence of these genes during evolution correlate with the acquisition of new functions in different species? By employing an evolutionary conservation analysis, the sequence of occurrence of biological processes during evolutionary history can be elucidated. Here we summarize and analyze the conservation status of the genes involved in sphingomyelin metabolism.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Huan Jiang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Moran Hu
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Yingyun Gong
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| | - Hongwen Zhou
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China
| |
Collapse
|
4
|
Bos JE, Duindam N, Kock TJF, Siegler MA, Wezenberg SJ. Control of Bilayer Transport through a Photoswitchable Membrane-Stiffening Agent. Angew Chem Int Ed Engl 2024:e202420232. [PMID: 39661481 DOI: 10.1002/anie.202420232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
The mobility of proteins in the bilayer membrane is affected by (local) changes in lipid environment, which is important to their biological functioning. Artificial molecular systems that-to some extent-imitate tasks of membrane-embedded proteins are increasingly developed, however, they are usually controlled through responsive units in their core structure. Here we present an alternative approach based on an amphiphilic stiff-stilbene derivative that enables control of membrane fluidity by light. The fluidity increase upon E-to-Z isomerization is shown to enhance the activity of known synthetic anion transporters as a result of a higher mobility. The photoisomerization process is studied by UV/Vis and 1H NMR spectroscopy in solution and in POPC vesicles, where the light-induced changes in fluidity and hence, activity of anion transporters, are monitored by fluorescence spectroscopy. Dynamic light-scattering (DLS) and cryo-EM studies show that vesicle integrity is not impaired by photoswitching. Our work introduces a versatile approach to control solute transport by carrier molecules. Moreover, the photocontrol over membrane fluidity and, with that, mobility could eventually be used for directed motion, which we expect to be key in achieving active transport in the future.
Collapse
Affiliation(s)
- Jasper E Bos
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The, Netherlands
| | - Nol Duindam
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The, Netherlands
| | - Thomas J F Kock
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The, Netherlands
| | - Maxime A Siegler
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD-21218, USA
| | - Sander J Wezenberg
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The, Netherlands
| |
Collapse
|
5
|
Müller GA, Müller TD. A "poly-matter network" conception of biological inheritance. Genetica 2024; 152:211-230. [PMID: 39425866 PMCID: PMC11541361 DOI: 10.1007/s10709-024-00216-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Here we intend to shift the "DNA- and information-centric" conception of biological inheritance, with the accompanying exclusion of any non-DNA matter, to a "poly-matter network" framework which, in addition to DNA, considers the action of other cellular membranous constituents. These cellular structures, in particular organelles and plasma membranes, express "landscapes" of specific topologies at their surfaces, which may become altered in response to certain environmental factors. These so-called "membranous environmental landscapes" (MELs), which replicate by self-organization / autopoiesis rather than self-assembly, are transferred from donor to acceptor cells by various - vesicular and non-vesicular - mechanisms and exert novel features in the acceptor cells. The "DNA-centric" conception may be certainly explanatorily sufficient for the transfer of heritable phenotype variation to acceptor cells following the copying of DNA in donor cells and thereby for the phenomenon of biological inheritance of traits. However, it is not causally sufficient. With the observation of phenotype variation, as initially manifested during bacterial transformation, the impact of environmental factors, such as nutrition and stress, in the differential regulation of gene expression has been widely accepted and resulted in intense efforts to resolve the underlying epigenetic mechanisms. However, these are explained under a conceptual frame where the DNA (and associated proteins) are the only matter of inheritance. In contrast, it is our argumentation that inheritance can only be adequately understood as the transfer of DNA in concert with non-DNA matter in a "poly-matter network" conception. The adequate inclusion of the transfer of non-DNA matter is still a desideratum of future genetic research, which may pave the way for the experimental elucidation not only of how DNA and membrane matter act in concert to enable the inheritance of innate traits, but also whether they interact for that of acquired biological traits. Moreover, the "poly-matter network" conception may open new perspectives for an understanding of the pathogenesis of "common complex" diseases.
Collapse
Affiliation(s)
- Günter A Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany.
- Biology and Technology Studies Institute Munich (BITSIM), Lappenweg 16, 80939, Munich, Germany.
- Media, Culture and Society, Department of Media Studies, Faculty of Arts and Humanities, University Paderborn, Warburger Str. 100, 33098, Paderborn, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
6
|
Bos J, Siegler MA, Wezenberg SJ. Activity Control of a Synthetic Transporter by Photodynamic Modulation of Membrane Mobility and Incorporation. J Am Chem Soc 2024; 146:31085-31093. [PMID: 39485737 PMCID: PMC11565646 DOI: 10.1021/jacs.4c10952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024]
Abstract
Artificial transmembrane transport systems are receiving a great deal of attention for their potential therapeutic application. A major challenge is to switch their activity in response to environmental stimuli, which has been achieved mostly by modulating the binding affinity. We demonstrate here that the activity of a synthetic anion transporter can be controlled through changes in the membrane mobility and incorporation. The transporters─equipped with azobenzene photoswitches─poorly incorporate into the bilayer membrane as their thermally stable (E,E,E)-isomers, but incorporation is triggered by UV irradiation to give the (Z)-containing isomers. The latter isomers, however, are found to have a lower mobility and are therefore the least active transporters. This opposite effect of E-Z isomerization on transport capability offers unique photocontrol as is demonstrated by in situ irradiation studies during the used transport assays. These results help to understand the behavior of artificial transporters in a bilayer and are highly important to future designs, with new modes of biological activity and with the possibility to direct motion, which may be crucial toward achieving active transport.
Collapse
Affiliation(s)
- Jasper
E. Bos
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Maxime A. Siegler
- Department
of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, Maryland 21218, United States
| | - Sander J. Wezenberg
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
7
|
Al Mismar R, Samavarchi-Tehrani P, Seale B, Kasmaeifar V, Martin CE, Gingras AC. Extracellular proximal interaction profiling by cell surface-targeted TurboID reveals LDLR as a partner of liganded EGFR. Sci Signal 2024; 17:eadl6164. [PMID: 39499777 DOI: 10.1126/scisignal.adl6164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 05/25/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024]
Abstract
Plasma membrane proteins play pivotal roles in receiving and transducing signals from other cells and from the environment and are vital for cellular functionality. Enzyme-based, proximity-dependent approaches, such as biotin identification (BioID), combined with mass spectrometry have begun to illuminate the landscape of proximal protein interactions within intracellular compartments. To extend the potential of these approaches to study the extracellular environment, we developed extracellular TurboID (ecTurboID), a method designed to profile the interactions between proteins on the surfaces of living cells over short timescales using the fast-acting biotin ligase TurboID. After optimizing our experimental and data analysis strategies to capture extracellular proximity interactions, we used ecTurboID to reveal the proximal interactomes of several plasma membrane proteins, including the epidermal growth factor receptor (EGFR). We found that EGF stimulation induced an association between EGFR and the low-density lipoprotein receptor (LDLR) and changed the interactome of LDLR by increasing its proximity with proteins that regulate EGFR signaling. The identification of this interaction between two well-studied and clinically relevant receptors illustrates the utility of our modified proximity labeling methodology for identifying dynamic extracellular associations between plasma membrane proteins.
Collapse
Affiliation(s)
- Rasha Al Mismar
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | | | - Brendon Seale
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Canada
| | - Vesal Kasmaeifar
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Claire E Martin
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
8
|
Shubhrasmita Sahu S, Sarkar P, Chattopadhyay A. Quantitation of F-actin in cytoskeletal reorganization: Context, methodology and implications. Methods 2024; 230:44-58. [PMID: 39074540 DOI: 10.1016/j.ymeth.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
The actin cytoskeleton is involved in a large number of cellular signaling events in addition to providing structural integrity to the cell. Actin polymerization is a key event during cellular signaling. Although the role of actin cytoskeleton in cellular processes such as trafficking and motility has been extensively studied, the reorganization of the actin cytoskeleton upon signaling has been rarely explored due to lack of suitable assays. Keeping in mind this lacuna, we developed a confocal microscopy based approach that relies on high magnification imaging of cellular F-actin, followed by image reconstruction using commercially available software. In this review, we discuss the context and relevance of actin quantitation, followed by a detailed hands-on approach of the methodology involved with specific points on troubleshooting and useful precautions. In the latter part of the review, we elucidate the method by discussing applications of actin quantitation from our work in several important problems in contemporary membrane biology ranging from pathogen entry into host cells, to GPCR signaling and membrane-cytoskeleton interaction. We envision that future discovery of cell-permeable novel fluorescent probes, in combination with genetically encoded actin-binding reporters, would allow real-time visualization of actin cytoskeleton dynamics to gain deeper insights into active cellular processes in health and disease.
Collapse
Affiliation(s)
- Subhashree Shubhrasmita Sahu
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Department of Biochemistry, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
9
|
Diao N, Hou J, Peng X, Wang Y, He A, Gao H, Yang L, Guo P, Wang J, Han D. Multiplexed and Quantitative Imaging of Live-Cell Membrane Proteins by a Precise and Controllable DNA-Encoded Amplification Reaction. Angew Chem Int Ed Engl 2024; 63:e202406330. [PMID: 38979704 DOI: 10.1002/anie.202406330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/10/2024]
Abstract
Amplifying DNA conjugated affinity ligands can improve the sensitivity and multiplicity of cell imaging and play a crucial role in comprehensively deciphering cellular heterogeneity and dynamic changes during development and disease. However, the development of one-step, controllable, and quantitative DNA amplification methods for multiplexed imaging of live-cell membrane proteins is challenging. Here, we introduce the template adhesion reaction (TAR) method for assembling amplifiable DNA sequences with different affinity ligands, such as aptamers or antibodies, for amplified and multiplexed imaging of live-cell membrane proteins with high quantitative fidelity. The precisely controllable TAR enables proportional amplification of membrane protein targets with variable abundances by modulating the concentration ratios of hairpin templates and primers, thus allowing sensitive visualization of multiple membrane proteins with enhanced signal-to-noise ratios (SNRs) without disturbing their original ratios. Using TAR, we achieved signal-enhanced imaging of six proteins on the same live-cell within 1-2 h. TAR represents an innovative and programmable molecular toolkit for multiplexed profiling of membrane proteins in live-cells.
Collapse
Affiliation(s)
- Nannan Diao
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jianing Hou
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xinyu Peng
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Life Science, Shanghai University, Shanghai, 200444, China
| | - Yaru Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Axin He
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Haiyan Gao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| | - Linlin Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Pei Guo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Junyan Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Da Han
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
10
|
Milne SM, Edeen PT, Fay DS. TAT-1, a phosphatidylserine flippase, affects molting and regulates membrane trafficking in the epidermis of C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613099. [PMID: 39314363 PMCID: PMC11419146 DOI: 10.1101/2024.09.15.613099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Membrane trafficking is a conserved process required for the movement and distribution of proteins and other macromolecules within cells. The Caenorhabditis elegans NIMA-related kinases NEKL-2 (human NEK8/9) and NEKL-3 (human NEK6/7) are conserved regulators of membrane trafficking and are required for the completion of molting. We used a genetic approach to identify reduction-of-function mutations in tat-1 that suppress nekl -associated molting defects. tat-1 encodes the C. elegans ortholog of mammalian ATP8A1/2, a phosphatidylserine (PS) flippase that promotes the asymmetric distribution of PS to the cytosolic leaflet of lipid membrane bilayers. CHAT-1 (human CDC50), a conserved chaperone, was required for the correct localization of TAT-1, and chat-1 inhibition strongly suppressed nekl defects. Using a PS sensor, we found that TAT-1 was required for the normal localization of PS at apical endosomes and that loss of TAT-1 led to aberrant endosomal morphologies. Consistent with this, TAT-1 localized to early endosomes and to recycling endosomes marked with RME-1, the C. elegans ortholog of the human EPS15 homology (EH) domain-containing protein, EHD1. TAT-1, PS biosynthesis, and the PS-binding protein RFIP-2 (human RAB11-FIP2) were all required for the normal localization of RME-1 to apical endosomes. Consistent with these proteins functioning together, inhibition of RFIP-2 or RME-1 led to the partial suppression of nekl molting defects, as did the inhibition of PS biosynthesis. Using the auxin-inducible degron system, we found that depletion of NEKL-2 or NEKL-3 led to defects in RME-1 localization and that a reduction in TAT-1 function partially restored RME-1 localization in NEKL-3-depleted cells. ARTICLE SUMMARY Endocytosis is an essential process required for the movement of proteins and lipids within cells. NEKL-2 and NEKL-3, two evolutionarily conserved proteins in the nematode Caenorhabditis elegans , are important regulators of endocytosis. In the current study, the authors describe a new functional link between the NEKLs and several proteins with known roles in endocytosis including TAT-1, a conserved enzyme that moves lipids between the bilayers of cellular membranes. As previous work implicated NEKLs in developmental defects and cancer, the present study can provide new insights into how the misregulation of endocytosis affects human health and disease.
Collapse
|
11
|
Barton MI, Paterson RL, Denham EM, Goyette J, van der Merwe PA. Ligand requirements for immunoreceptor triggering. Commun Biol 2024; 7:1138. [PMID: 39271744 PMCID: PMC11399299 DOI: 10.1038/s42003-024-06817-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Leukocytes interact with other cells using cell surface receptors. The largest group of such receptors are non-catalytic tyrosine phosphorylated receptors (NTRs), also called immunoreceptors. NTR signalling requires phosphorylation of cytoplasmic tyrosine residues by SRC-family tyrosine kinases. How ligand binding to NTRs induces this phosphorylation, also called NTR triggering, remains controversial, with roles suggested for size-based segregation, clustering, and mechanical force. Here we exploit a recently developed cell-surface generic ligand system to explore the ligand requirements for NTR triggering. We examine the effect of varying the ligand's length, mobility and valency on the activation of representative members of four NTR families: SIRPβ1, Siglec 14, NKp44 and TREM-1. Increasing the ligand length impairs activation via NTRs, despite enhancing cell-cell conjugation, while varying ligand mobility has little effect on either conjugation or activation. Increasing the valency of the ligand, while enhancing cell-cell conjugation, does not enhance activation at equivalent levels of conjugation. These findings are more consistent with a role for size-based segregation, rather than mechanical force or clustering, in NTR triggering, suggesting a role for the kinetic-segregation model.
Collapse
Affiliation(s)
- Michael I Barton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Rachel L Paterson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Parque de Ciência e Tecnologia Avepark, Zona Industrial da Gandra, Barco, Portugal
| | - Eleanor M Denham
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Enara Bio, The Magdalen Centre, Oxford Science Park, 1 Robert Robinson Avenue, Oxford, UK
| | - Jesse Goyette
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Department of Molecular Medicine, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
12
|
Tuomivaara ST, Teo CF, Jan YN, Wiita AP, Jan LY. SLAPSHOT reveals rapid dynamics of extracellularly exposed proteome in response to calcium-activated plasma membrane phospholipid scrambling. Commun Biol 2024; 7:1060. [PMID: 39210032 PMCID: PMC11362511 DOI: 10.1038/s42003-024-06729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
To facilitate our understanding of proteome dynamics during signaling events, robust workflows affording fast time resolution without confounding factors are essential. We present Surface-exposed protein Labeling using PeroxidaSe, H2O2, and Tyramide-derivative (SLAPSHOT) to label extracellularly exposed proteins in a rapid, specific, and sensitive manner. Simple and flexible SLAPSHOT utilizes recombinant soluble APEX2 protein applied to cells, thus circumventing the engineering of tools and cells, biological perturbations, and labeling biases. We applied SLAPSHOT and quantitative proteomics to examine the TMEM16F-dependent plasma membrane remodeling in WT and TMEM16F KO cells. Time-course data ranging from 1 to 30 min of calcium stimulation revealed co-regulation of known protein families, including the integrin and ICAM families, and identified proteins known to reside in intracellular organelles as occupants of the freshly deposited extracellularly exposed membrane. Our data provide the first accounts of the immediate consequences of calcium signaling on the extracellularly exposed proteome.
Collapse
Affiliation(s)
- Sami T Tuomivaara
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences and Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, CA, USA
| | - Chin Fen Teo
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
- Department of Physiology, University of California, San Francisco, CA, USA
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
- Department of Physiology, University of California, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA.
| | - Lily Y Jan
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.
- Department of Physiology, University of California, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
| |
Collapse
|
13
|
Oslund RC, Holland PM, Lesley SA, Fadeyi OO. Therapeutic potential of cis-targeting bispecific antibodies. Cell Chem Biol 2024; 31:1473-1489. [PMID: 39111317 DOI: 10.1016/j.chembiol.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/13/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024]
Abstract
The growing clinical success of bispecific antibodies (bsAbs) has led to rapid interest in leveraging dual targeting in order to generate novel modes of therapeutic action beyond mono-targeting approaches. While bsAbs that bind targets on two different cells (trans-targeting) are showing promise in the clinic, the co-targeting of two proteins on the same cell surface through cis-targeting bsAbs (cis-bsAbs) is an emerging strategy to elicit new functionalities. This includes the ability to induce proximity, enhance binding to a target, increase target/cell selectivity, and/or co-modulate function on the cell surface with the goal of altering, reversing, or eradicating abnormal cellular activity that contributes to disease. In this review, we focus on the impact of cis-bsAbs in the clinic, their emerging applications, and untangle the intricacies of improving bsAb discovery and development.
Collapse
|
14
|
Tsai FC, Guérin G, Pernier J, Bassereau P. Actin-membrane linkers: Insights from synthetic reconstituted systems. Eur J Cell Biol 2024; 103:151402. [PMID: 38461706 DOI: 10.1016/j.ejcb.2024.151402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/10/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
At the cell surface, the actin cytoskeleton and the plasma membrane interact reciprocally in a variety of processes related to the remodeling of the cell surface. The actin cytoskeleton has been known to modulate membrane organization and reshape the membrane. To this end, actin-membrane linking molecules play a major role in regulating actin assembly and spatially direct the interaction between the actin cytoskeleton and the membrane. While studies in cells have provided a wealth of knowledge on the molecular composition and interactions of the actin-membrane interface, the complex molecular interactions make it challenging to elucidate the precise actions of the actin-membrane linkers at the interface. Synthetic reconstituted systems, consisting of model membranes and purified proteins, have been a powerful approach to elucidate how actin-membrane linkers direct actin assembly to drive membrane shape changes. In this review, we will focus only on several actin-membrane linkers that have been studied by using reconstitution systems. We will discuss the design principles of these reconstitution systems and how they have contributed to the understanding of the cellular functions of actin-membrane linkers. Finally, we will provide a perspective on future research directions in understanding the intricate actin-membrane interaction.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| | - Gwendal Guérin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France
| | - Julien Pernier
- Tumor Cell Dynamics Unit, Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif 94800, France
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| |
Collapse
|
15
|
Lüchtefeld I, Pivkin IV, Gardini L, Zare-Eelanjegh E, Gäbelein C, Ihle SJ, Reichmuth AM, Capitanio M, Martinac B, Zambelli T, Vassalli M. Dissecting cell membrane tension dynamics and its effect on Piezo1-mediated cellular mechanosensitivity using force-controlled nanopipettes. Nat Methods 2024; 21:1063-1073. [PMID: 38802520 PMCID: PMC11166569 DOI: 10.1038/s41592-024-02277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
The dynamics of cellular membrane tension and its role in mechanosensing, which is the ability of cells to respond to physical stimuli, remain incompletely understood, mainly due to the lack of appropriate tools. Here, we report a force-controlled nanopipette-based method that combines fluidic force microscopy with fluorescence imaging for precise manipulation of the cellular membrane tension while monitoring the impact on single-cell mechanosensitivity. The force-controlled nanopipette enables control of the indentation force imposed on the cell cortex as well as of the aspiration pressure applied to the plasma membrane. We show that this setup can be used to concurrently monitor the activation of Piezo1 mechanosensitive ion channels via calcium imaging. Moreover, the spatiotemporal behavior of the tension propagation is assessed with the fluorescent membrane tension probe Flipper-TR, and further dissected using molecular dynamics modeling. Finally, we demonstrate that aspiration and indentation act independently on the cellular mechanobiological machinery, that indentation induces a local pre-tension in the membrane, and that membrane tension stays confined by links to the cytoskeleton.
Collapse
Affiliation(s)
- Ines Lüchtefeld
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Igor V Pivkin
- Institute of Computing, Università della Svizzera Italiana, Lugano, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Lucia Gardini
- National Institute of Optics, National Research Council, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
| | | | | | - Stephan J Ihle
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Andreas M Reichmuth
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Marco Capitanio
- European Laboratory for Non-Linear Spectroscopy, University of Florence, Florence, Italy
- Physics and Astronomy Department, University of Florence, Florence, Italy
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Tomaso Zambelli
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland.
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK.
| |
Collapse
|
16
|
Li M, Xing X, Yuan J, Zeng Z. Research progress on the regulatory role of cell membrane surface tension in cell behavior. Heliyon 2024; 10:e29923. [PMID: 38720730 PMCID: PMC11076917 DOI: 10.1016/j.heliyon.2024.e29923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
Cell membrane surface tension has emerged as a pivotal biophysical factor governing cell behavior and fate. This review systematically delineates recent advances in techniques for cell membrane surface tension quantification, mechanosensing mechanisms, and regulatory roles of cell membrane surface tension in modulating major cellular processes. Micropipette aspiration, tether pulling, and newly developed fluorescent probes enable the measurement of cell membrane surface tension with spatiotemporal precision. Cells perceive cell membrane surface tension via conduits including mechanosensitive ion channels, curvature-sensing proteins (e.g. BAR domain proteins), and cortex-membrane attachment proteins (e.g. ERM proteins). Through membrane receptors like integrins, cells convert mechanical cues into biochemical signals. This conversion triggers cytoskeletal remodeling and extracellular matrix interactions in response to environmental changes. Elevated cell membrane surface tension suppresses cell spreading, migration, and endocytosis while facilitating exocytosis. Moreover, reduced cell membrane surface tension promotes embryonic stem cell differentiation and cancer cell invasion, underscoring cell membrane surface tension as a regulator of cell plasticity. Outstanding questions remain regarding cell membrane surface tension regulatory mechanisms and roles in tissue development/disease in vivo. Emerging tools to manipulate cell membrane surface tension with high spatiotemporal control in combination with omics approaches will facilitate the elucidation of cell membrane surface tension-mediated effects on signaling networks across various cell types/states. This will accelerate the development of cell membrane surface tension-based biomarkers and therapeutics for regenerative medicine and cancer. Overall, this review provides critical insights into cell membrane surface tension as a potent orchestrator of cell function, with broader impacts across mechanobiology.
Collapse
Affiliation(s)
- Manqing Li
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Xiumei Xing
- School of Public Health, Sun Yat-sen University, Guangzhou, 5180080, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518054, China
| | - Zhuoying Zeng
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518035, China
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| |
Collapse
|
17
|
Voce N, Stevenson P. Experimentally Probing the Effect of Confinement Geometry on Lipid Diffusion. J Phys Chem B 2024; 128:4404-4413. [PMID: 38574293 PMCID: PMC11089508 DOI: 10.1021/acs.jpcb.3c07388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
The lateral mobility of molecules within the cell membrane is ultimately governed by the local environment of the membrane. Confined regions induced by membrane structures, such as protein aggregates or the actin meshwork, occur over a wide range of length scales and can impede or steer the diffusion of membrane components. However, a detailed picture of the origins and nature of these confinement effects remains elusive. Here, we prepare model lipid systems on substrates patterned with confined domains of varying geometries constructed with different materials to explore the influences of physical boundary conditions and specific molecular interactions on diffusion. We demonstrate a platform that is capable of significantly altering and steering the long-range diffusion of lipids by using simple oxide deposition approaches, enabling us to systematically explore how confinement size and shape impact diffusion over multiple length scales. While we find that a "boundary condition" description of the system captures underlying trends in some cases, we are also able to directly compare our systems to analytical models, revealing the unexpected breakdown of several approximate solutions. Our results highlight the importance of considering the length scale dependence when discussing properties such as diffusion.
Collapse
Affiliation(s)
- Nicole Voce
- Department of Physics, Northeastern University, Boston, Massachusetts 02115, United States
| | - Paul Stevenson
- Department of Physics, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
18
|
Woolley L, Burbidge A, Vermant J, Christakopoulos F. A microrheological examination of insulin-secreting β-cells in healthy and diabetic-like conditions. SOFT MATTER 2024; 20:3464-3472. [PMID: 38573072 DOI: 10.1039/d3sm01141k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Pancreatic β-cells regulate glucose homeostasis through glucose-stimulated insulin secretion, which is hindered in type-2 diabetes. Transport of the insulin vesicles is expected to be affected by changes in the viscoelastic and transport properties of the cytoplasm. These are evaluated in situ through particle-tracking measurements using a rat insulinoma β-cell line. The use of inert probes assists in decoupling the material properties of the cytoplasm from the active transport through cellular processes. The effect of glucose-stimulated insulin secretion is examined, and the subsequent remodeling of the cytoskeleton, at constant effects of cell activity, is shown to result in reduced mobility of the tracer particles. Induction of diabetic-like conditions is identified to alter the mean-squared displacement of the passive particles in the cytoplasm and diminish its reaction to glucose stimulation.
Collapse
Affiliation(s)
- Lukas Woolley
- Department of Materials, ETH Zurich, Vladimir-Prelog-Weg 5, 8093 Zurich, Switzerland.
| | - Adam Burbidge
- Nestlé Research, Route de Jorat 57, vers-chez-les Blanc, 1000 Lausanne, Switzerland
| | - Jan Vermant
- Department of Materials, ETH Zurich, Vladimir-Prelog-Weg 5, 8093 Zurich, Switzerland.
| | - Fotis Christakopoulos
- Department of Materials, ETH Zurich, Vladimir-Prelog-Weg 5, 8093 Zurich, Switzerland.
| |
Collapse
|
19
|
Gryczak DW, Lenzi EK, Rosseto MP, Evangelista LR, Zola RS. Non-Markovian Diffusion and Adsorption-Desorption Dynamics: Analytical and Numerical Results. ENTROPY (BASEL, SWITZERLAND) 2024; 26:294. [PMID: 38667848 PMCID: PMC11048754 DOI: 10.3390/e26040294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
The interplay of diffusion with phenomena like stochastic adsorption-desorption, absorption, and reaction-diffusion is essential for life and manifests in diverse natural contexts. Many factors must be considered, including geometry, dimensionality, and the interplay of diffusion across bulk and surfaces. To address this complexity, we investigate the diffusion process in heterogeneous media, focusing on non-Markovian diffusion. This process is limited by a surface interaction with the bulk, described by a specific boundary condition relevant to systems such as living cells and biomaterials. The surface can adsorb and desorb particles, and the adsorbed particles may undergo lateral diffusion before returning to the bulk. Different behaviors of the system are identified through analytical and numerical approaches.
Collapse
Affiliation(s)
| | - Ervin K. Lenzi
- Departamento de Física, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil;
| | - Michely P. Rosseto
- Departamento de Física, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil;
| | - Luiz R. Evangelista
- Departamento de Física, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil;
- Istituto dei Sistemi Complessi (ISC–CNR), Via dei Taurini, 19, 00185 Rome, Italy
- Department of Molecular Science and Nanosystems, Ca’ Foscari University of Venice, Via Torino 155, 30175 Mestre (VE), Italy
| | - Rafael S. Zola
- Department of Physics, Universidade Tecnológica Federal do Paraná, Apucarana 86812-460, PR, Brazil;
| |
Collapse
|
20
|
Müller GA, Müller TD. Transfer of membrane(s) matter(s)-non-genetic inheritance of (metabolic) phenotypes? Front Mol Biosci 2024; 11:1347397. [PMID: 38516184 PMCID: PMC10955475 DOI: 10.3389/fmolb.2024.1347397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/26/2024] [Indexed: 03/23/2024] Open
Abstract
Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are anchored at the outer phospholipid layer of eukaryotic plasma membranes exclusively by a glycolipid. GPI-APs are not only released into extracellular compartments by lipolytic cleavage. In addition, certain GPI-APs with the glycosylphosphatidylinositol anchor including their fatty acids remaining coupled to the carboxy-terminus of their protein components are also detectable in body fluids, in response to certain stimuli, such as oxidative stress, radicals or high-fat diet. As a consequence, the fatty acid moieties of GPI-APs must be shielded from access of the aqueous environment by incorporation into membranes of extracellular vesicles or into micelle-like complexes together with (lyso)phospholipids and cholesterol. The GPI-APs released from somatic cells and tissues are transferred via those complexes or EVs to somatic as well as pluripotent stem cells with metabolic consequences, such as upregulation of glycogen and lipid synthesis. From these and additional findings, the following hypotheses are developed: i) Transfer of GPI-APs via EVs or micelle-like complexes leads to the induction of new phenotypes in the daughter cells or zygotes, which are presumably not restricted to metabolism. ii) The membrane topographies transferred by the concerted action of GPI-APs and interacting components are replicated by self-organization and self-templation and remain accessible to structural changes by environmental factors. iii) Transfer from mother cells and gametes to their daughter cells and zygotes, respectively, is not restricted to DNA and genes, but also encompasses non-genetic matter, such as GPI-APs and specific membrane constituents. iv) The intergenerational transfer of membrane matter between mammalian organisms is understood as an epigenetic mechanism for phenotypic plasticity, which does not rely on modifications of DNA and histones, but is regarded as molecular mechanism for the inheritance of acquired traits, such as complex metabolic diseases. v) The missing interest in research of non-genetic matter of inheritance, which may be interpreted in the sense of Darwin's "Gemmules" or Galton's "Stirps", should be addressed in future investigations of the philosophy of science and sociology of media.
Collapse
Affiliation(s)
- Günter A. Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Oberschleissheim, Germany
- Department of Media Studies, Media, Culture and Society, Faculty of Arts and Humanities, University Paderborn, Paderborn, Germany
| | - Timo D. Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), Oberschleissheim, Germany
| |
Collapse
|
21
|
Kumar V, Shepard Bryan J, Rojewski A, Manzo C, Pressé S. Learning Continuous 2D Diffusion Maps from Particle Trajectories without Data Binning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582378. [PMID: 38464131 PMCID: PMC10925201 DOI: 10.1101/2024.02.27.582378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Diffusion coefficients often vary across regions, such as cellular membranes, and quantifying their variation can provide valuable insight into local membrane properties such as composition and stiffness. Toward quantifying diffusion coefficient spatial maps and uncertainties from particle tracks, we use a Bayesian method and place Gaussian Process (GP) Priors on the maps. For the sake of computational efficiency, we leverage inducing point methods on GPs arising from the mathematical structure of the data giving rise to non-conjugate likelihood-prior pairs. We analyze both synthetic data, where ground truth is known, as well as data drawn from live-cell single-molecule imaging of membrane proteins. The resulting tool provides an unsupervised method to rigorously map diffusion coefficients continuously across membranes without data binning.
Collapse
Affiliation(s)
- Vishesh Kumar
- Center for Biological Physics, Arizona State University, USA
- Department of Physics, Arizona State University, USA
| | - J. Shepard Bryan
- Center for Biological Physics, Arizona State University, USA
- Department of Physics, Arizona State University, USA
| | - Alex Rojewski
- Center for Biological Physics, Arizona State University, USA
- Department of Physics, Arizona State University, USA
| | - Carlo Manzo
- Facultat de Ciéncies, Tecnologia i Enginyeries, Universitat de Vic – Universitat Central de Catalunya (UVic-UCC), C. de la Laura,13, 08500 Vic, Barcelona, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500 Vic, Barcelona, Spain
| | - Steve Pressé
- Center for Biological Physics, Arizona State University, USA
- Department of Physics, Arizona State University, USA
- School of Molecular Sciences, Arizona State University
| |
Collapse
|
22
|
Zhang K, Nie Q, Chi-Kong Lau T, Kit Kwok C. Rational Design of L-RNA Aptamer-Peptide Conjugate for Efficient Cell Uptake and G-quadruplex-Mediated Gene Control. Angew Chem Int Ed Engl 2024; 63:e202310798. [PMID: 38156978 DOI: 10.1002/anie.202310798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/19/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
RNA G-quadruplexes (D-rG4s) are prevalent in the transcriptome and play crucial regulatory roles in various biological processes. Recently, L-RNA aptamers have been reported to recognize functional rG4s with a strong binding affinity and specificity. However, owing to the poor cell penetration capacity of L-RNA aptamers, their biological applications are currently limited. Herein, we rationally design an L-RNA aptamer-peptide conjugate, Tamra_Ahx_R8_L-Apt.4-1c, which can efficiently translocate into the cytosol and target the rG4 of interest. Notably, we demonstrate diverse regulatory roles of Tamra_Ahx_R8_L-Apt.4-1c on rG4 motif present in different regions of mRNAs and further expand the application in different cell lines. Our novel and biocompatible conjugate enhances the cellular uptake of the L-RNA aptamer, and our robust strategy enables non-canonical RNA structures to be targeted by L-RNA aptamers for gene control in cells.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Chemistry and State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qichang Nie
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Terrence Chi-Kong Lau
- Shenzhen Research Institute of, City University of Hong Kong, Shenzhen, 518057, China
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chun Kit Kwok
- Department of Chemistry and State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong SAR, 999077, China
- Shenzhen Research Institute of, City University of Hong Kong, Shenzhen, 518057, China
| |
Collapse
|
23
|
Hermann DM, Peruzzotti-Jametti L, Giebel B, Pluchino S. Extracellular vesicles set the stage for brain plasticity and recovery by multimodal signalling. Brain 2024; 147:372-389. [PMID: 37768167 PMCID: PMC10834259 DOI: 10.1093/brain/awad332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are extremely versatile naturally occurring membrane particles that convey complex signals between cells. EVs of different cellular sources are capable of inducing striking therapeutic responses in neurological disease models. Differently from pharmacological compounds that act by modulating defined signalling pathways, EV-based therapeutics possess multiple abilities via a variety of effectors, thus allowing the modulation of complex disease processes that may have very potent effects on brain tissue recovery. When applied in vivo in experimental models of neurological diseases, EV-based therapeutics have revealed remarkable effects on immune responses, cell metabolism and neuronal plasticity. This multimodal modulation of neuroimmune networks by EVs profoundly influences disease processes in a highly synergistic and context-dependent way. Ultimately, the EV-mediated restoration of cellular functions helps to set the stage for neurological recovery. With this review we first outline the current understanding of the mechanisms of action of EVs, describing how EVs released from various cellular sources identify their cellular targets and convey signals to recipient cells. Then, mechanisms of action applicable to key neurological conditions such as stroke, multiple sclerosis and neurodegenerative diseases are presented. Pathways that deserve attention in specific disease contexts are discussed. We subsequently showcase considerations about EV biodistribution and delineate genetic engineering strategies aiming at enhancing brain uptake and signalling. By sketching a broad view of EV-orchestrated brain plasticity and recovery, we finally define possible future clinical EV applications and propose necessary information to be provided ahead of clinical trials. Our goal is to provide a steppingstone that can be used to critically discuss EVs as next generation therapeutics for brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
24
|
Barrantes FJ. Modulation of a rapid neurotransmitter receptor-ion channel by membrane lipids. Front Cell Dev Biol 2024; 11:1328875. [PMID: 38274273 PMCID: PMC10808158 DOI: 10.3389/fcell.2023.1328875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Membrane lipids modulate the proteins embedded in the bilayer matrix by two non-exclusive mechanisms: direct or indirect. The latter comprise those effects mediated by the physicochemical state of the membrane bilayer, whereas direct modulation entails the more specific regulatory effects transduced via recognition sites on the target membrane protein. The nicotinic acetylcholine receptor (nAChR), the paradigm member of the pentameric ligand-gated ion channel (pLGIC) superfamily of rapid neurotransmitter receptors, is modulated by both mechanisms. Reciprocally, the nAChR protein exerts influence on its surrounding interstitial lipids. Folding, conformational equilibria, ligand binding, ion permeation, topography, and diffusion of the nAChR are modulated by membrane lipids. The knowledge gained from biophysical studies of this prototypic membrane protein can be applied to other neurotransmitter receptors and most other integral membrane proteins.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Biomedical Research Institute (BIOMED), Catholic University of Argentina (UCA)–National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
25
|
Sampieri A, Padilla-Flores T, Thawani AR, Lam PY, Fuchter MJ, Peterson R, Vaca L. The conducting state of TRPA1 modulates channel lateral mobility. Cell Calcium 2023; 116:102800. [PMID: 37776645 DOI: 10.1016/j.ceca.2023.102800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/04/2023] [Accepted: 09/16/2023] [Indexed: 10/02/2023]
Abstract
We have studied Danio rerio (Zebrafish) TRPA1 channel using a method that combines single channel electrophysiological and optical recordings to evaluate lateral mobility and channel gating simultaneously in single channels. TRPA1 channel activation by two distinct chemical ligands: allyl isothiocyanate (AITC) and TRPswitch B, results in substantial reduction of channel lateral mobility at the plasma membrane. Incubation with the cholesterol sequestering agent methyl-β-cyclodextrin (MβCD), prevents the reduction on lateral mobility induced by the two chemical agonists. This results strongly suggest that the open conformation of TRPA1 modulates channel lateral mobility probably by facilitating the insertion of the channel into cholesterol-enriched domains at the plasma membrane.
Collapse
Affiliation(s)
- Alicia Sampieri
- Instituto de Fisiología Celular. Departamento de Biología Celular y del desarrollo. Universidad Nacional Autónoma de México. México, CDMX 04510, Mexico
| | - Teresa Padilla-Flores
- Instituto de Fisiología Celular. Departamento de Biología Celular y del desarrollo. Universidad Nacional Autónoma de México. México, CDMX 04510, Mexico
| | - Aditya R Thawani
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, London W12 OBZ, United Kingdom
| | - Pui-Ying Lam
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, Wisconsin 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, Wisconsin, 53226, USA
| | - Matthew J Fuchter
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, London W12 OBZ, United Kingdom
| | - Randall Peterson
- College of Pharmacy, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
| | - Luis Vaca
- Instituto de Fisiología Celular. Departamento de Biología Celular y del desarrollo. Universidad Nacional Autónoma de México. México, CDMX 04510, Mexico.
| |
Collapse
|
26
|
Azbazdar Y, Demirci Y, Heger G, Ipekgil D, Karabicici M, Ozhan G. Comparative membrane lipidomics of hepatocellular carcinoma cells reveals diacylglycerol and ceramide as key regulators of Wnt/β-catenin signaling and tumor growth. Mol Oncol 2023; 17:2314-2336. [PMID: 37699867 PMCID: PMC10620124 DOI: 10.1002/1878-0261.13520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/22/2023] [Accepted: 09/09/2023] [Indexed: 09/14/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is largely associated with aberrant activation of Wnt/β-catenin signaling. Nevertheless, how membrane lipid composition is altered in HCC cells with abnormal Wnt signaling remains elusive. Here, by exploiting comprehensive lipidome profiling, we unravel the membrane lipid composition of six different HCC cell lines with mutations in components of Wnt/β-catenin signaling, leading to differences in their endogenous signaling activity. Among the differentially regulated lipids are diacylglycerol (DAG) and ceramide, which were downregulated at the membrane of HCC cells after Wnt3a treatment. DAG and ceramide enhanced Wnt/β-catenin signaling by inducing caveolin-mediated endocytosis of the canonical Wnt-receptor complex, while their depletion suppressed the signaling activity along with a reduction of caveolin-mediated endocytosis in SNU475 and HepG2 cells. Moreover, depletion of DAG and ceramide significantly impeded the proliferation, tumor growth, and in vivo migration capacity of SNU475 and HepG2 cells. This study, by pioneering plasma membrane lipidome profiling in HCC cells, exhibits the remarkable potential of lipids to correct dysregulated signaling pathways in cancer and stop abnormal tumor growth.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG)Dokuz Eylul University Health CampusIzmirTurkey
- Izmir International Biomedicine and Genome Institute (IBG‐Izmir)Dokuz Eylul UniversityIzmirTurkey
- Present address:
Department of Biological ChemistryUniversity of California Los AngelesCAUSA
| | - Yeliz Demirci
- Izmir Biomedicine and Genome Center (IBG)Dokuz Eylul University Health CampusIzmirTurkey
- Izmir International Biomedicine and Genome Institute (IBG‐Izmir)Dokuz Eylul UniversityIzmirTurkey
- Present address:
Wellcome Sanger InstituteCambridgeUK
| | | | - Dogac Ipekgil
- Izmir Biomedicine and Genome Center (IBG)Dokuz Eylul University Health CampusIzmirTurkey
- Izmir International Biomedicine and Genome Institute (IBG‐Izmir)Dokuz Eylul UniversityIzmirTurkey
| | - Mustafa Karabicici
- Izmir Biomedicine and Genome Center (IBG)Dokuz Eylul University Health CampusIzmirTurkey
- Izmir International Biomedicine and Genome Institute (IBG‐Izmir)Dokuz Eylul UniversityIzmirTurkey
- Present address:
Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG)Dokuz Eylul University Health CampusIzmirTurkey
- Izmir International Biomedicine and Genome Institute (IBG‐Izmir)Dokuz Eylul UniversityIzmirTurkey
- Present address:
Department of Molecular Biology and GeneticsIzmir Institute of TechnologyTurkey
| |
Collapse
|
27
|
Sun H, Qi H, Hu W, Guan L, Xue J, Ai Y, Wang Y, Ding M, Liang Q. Single Nanovesicles Tracking Reveals Their Heterogeneous Extracellular Adsorptions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301888. [PMID: 37467296 DOI: 10.1002/smll.202301888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/07/2023] [Indexed: 07/21/2023]
Abstract
The vigorous nanomedicine offers significant possibilities for effective therapeutics of various diseases, and nanovesicles (NVs) represented by artificial liposomes and natural exosomes and cytomembranes especially show great potential. However, their complex interactions with cells, particularly the heterogeneous extracellular adsorptions, are difficult to analyze spatiotemporally due to the transient dynamics. In this study, by single NVs tracking, the extracellular NVs adsorptions are directly observed and their heterogeneous characteristics are revealed. Briefly, plenty of NVs adsorbed on HCT116 cells are tracked and classified, and it is discovered that they exhibit various diffusion properties from different extracellular regions: stable adsorptions on the rear surface and restricted adsorptions on the front protrusion. After the hydrolysis of hyaluronic acid in the extracellular matrix by hyaluronidase, the restricted adsorptions are further weakened and manifested as dissociative adsorptions, which demonstrated reduced total NVs adsorptions from a single-cell and single-particle perspective. Compared with traditional static analysis, the spatiotemporal tracking and heterogeneous results not only reveal the extracellular NVs-cell interactions but also inspire a wide variety of nanomedicine and their nano-investigations.
Collapse
Affiliation(s)
- Hua Sun
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Huibo Qi
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Liandi Guan
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jianfeng Xue
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Mingyu Ding
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
28
|
Dibsy R, Bremaud E, Mak J, Favard C, Muriaux D. HIV-1 diverts cortical actin for particle assembly and release. Nat Commun 2023; 14:6945. [PMID: 37907528 PMCID: PMC10618566 DOI: 10.1038/s41467-023-41940-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
Enveloped viruses assemble and bud from the host cell membranes. Any role of cortical actin in these processes have often been a source of debate. Here, we assessed if cortical actin was involved in HIV-1 assembly in infected CD4 T lymphocytes. Our results show that preventing actin branching not only increases HIV-1 particle release but also the number of individual HIV-1 Gag assembly clusters at the T cell plasma membrane. Indeed, in infected T lymphocytes and in in vitro quantitative model systems, we show that HIV-1 Gag protein prefers areas deficient in F-actin for assembling. Finally, we found that the host factor Arpin, an inhibitor of Arp2/3 branched actin, is recruited at the membrane of infected T cells and it can associate with the viral Gag protein. Altogether, our data show that, for virus assembly and particle release, HIV-1 prefers low density of cortical actin and may favor local actin debranching by subverting Arpin.
Collapse
Affiliation(s)
- Rayane Dibsy
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Erwan Bremaud
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Johnson Mak
- Institute for Glycomics, Griffith University, Brisbane, Australia
| | - Cyril Favard
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Delphine Muriaux
- Institute of Research in Infectious disease of Montpellier (IRIM), University of Montpellier, UMR9004 CNRS, Montpellier, France.
| |
Collapse
|
29
|
Mayor S, Bhat A, Kusumi A. A Survey of Models of Cell Membranes: Toward a New Understanding of Membrane Organization. Cold Spring Harb Perspect Biol 2023; 15:a041394. [PMID: 37643877 PMCID: PMC10547391 DOI: 10.1101/cshperspect.a041394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The cell membrane, the boundary that separates living cells from their environment, has been the subject of study for over a century. The fluid-mosaic model of Singer and Nicolson in 1972 proposed the plasma membrane as a two-dimensional fluid composed of lipids and proteins. Fifty years hence, advances in biophysical and biochemical tools, particularly optical imaging techniques, have allowed for a better understanding of the physical nature, organization, and composition of cell membranes. This has been made possible by visualizing membrane heterogeneities and their dynamics and appreciating the asymmetrical arrangement of lipids in living cell membranes. Despite these advances, mechanisms underlying the local spatiotemporal organization of membrane components remain unclear. This review surveys various models of membrane organization, culminating in a new model that incorporates nonequilibrium processes and forces exerted by interactions with extramembrane elements such as the actin cytoskeleton. The proposed model provides a comprehensive understanding of membrane organization, taking into account the dynamic nature of the cell membrane and its interactions with its immediate environment.
Collapse
Affiliation(s)
- Satyajit Mayor
- National Centre for Biological Science, TIFR, Bangalore 560065, India
| | - Abrar Bhat
- National Centre for Biological Science, TIFR, Bangalore 560065, India
| | - Akihiro Kusumi
- Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
30
|
Hahn KM, Itano MS, Loew LM, Vitriol EA. Celebrating the creative scientific life of Ken Jacobson. Biophys J 2023; 122:E1-E4. [PMID: 37643609 PMCID: PMC10541490 DOI: 10.1016/j.bpj.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023] Open
Affiliation(s)
- Klaus M Hahn
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michelle S Itano
- Department of Cell Biology & Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Leslie M Loew
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Eric A Vitriol
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia.
| |
Collapse
|
31
|
Dasgupta A, Ngo HT, Tschoerner D, Touret N, da Rocha-Azevedo B, Jaqaman K. Multiscale imaging and quantitative analysis of plasma membrane protein-cortical actin interplay. Biophys J 2023; 122:3798-3815. [PMID: 37571825 PMCID: PMC10541498 DOI: 10.1016/j.bpj.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/19/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023] Open
Abstract
The spatiotemporal organization of cell surface receptors is important for cell signaling. Cortical actin (CA), the subset of the actin cytoskeleton subjacent to the plasma membrane (PM), plays a large role in cell surface receptor organization. However, this has been shown largely through actin perturbation experiments, which raise concerns of nonspecific effects and preclude quantification of actin architecture and dynamics under unperturbed conditions. These limitations make it challenging to predict how changes in CA properties can affect receptor organization. To derive direct relationships between the architecture and dynamics of CA and the spatiotemporal organization of PM proteins, including cell surface receptors, we developed a multiscale imaging and computational analysis framework based on the integration of single-molecule imaging (SMI) of PM proteins and fluorescent speckle microscopy (FSM) of CA (combined: SMI-FSM) in the same live cell. SMI-FSM revealed differential relationships between PM proteins and CA based on the PM proteins' actin binding ability, diffusion type, and local CA density. Combining SMI-FSM with subcellular region analysis revealed differences in CA dynamics that were predictive of differences in PM protein mobility near ruffly cell edges versus closer to the cell center. SMI-FSM also highlighted the complexity of cell-wide actin perturbation, where we found that global changes in actin properties caused by perturbation were not necessarily reflected in the CA properties near PM proteins, and that the changes in PM protein properties upon perturbation varied based on the local CA environment. Given the widespread use of SMI as a method to study the spatiotemporal organization of PM proteins and the versatility of SMI-FSM, we expect it to be widely applicable to enable future investigation of the influence of CA architecture and dynamics on different PM proteins, especially in the context of actin-dependent cellular processes.
Collapse
Affiliation(s)
- Aparajita Dasgupta
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Huong-Tra Ngo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Deryl Tschoerner
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nicolas Touret
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Bruno da Rocha-Azevedo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Khuloud Jaqaman
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
32
|
Mogilner A, Savinov M. Crawling, waving, inch worming, dilating, and pivoting mechanics of migrating cells: Lessons from Ken Jacobson. Biophys J 2023; 122:3551-3559. [PMID: 36934300 PMCID: PMC10541468 DOI: 10.1016/j.bpj.2023.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Research on the locomotion of single cells on hard, flat surfaces brought insight into the mechanisms of leading-edge protrusion, spatially graded adhesion, front-rear coordination, and how intracellular and traction forces are harnessed to execute various maneuvers. Here, we highlight how, by studying a variety of cell types, shapes, and movements, Ken Jacobson and his collaborators made several discoveries that triggered the mechanistic understanding of cell motility. We then review the recent advancements and current perspectives in this field.
Collapse
Affiliation(s)
- Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, New York; Department of Biology, New York University, New York, New York.
| | - Mariya Savinov
- Courant Institute of Mathematical Sciences, New York University, New York, New York
| |
Collapse
|
33
|
Frey K, Rohrer L, Frommelt F, Ringwald M, Potapenko A, Goetze S, von Eckardstein A, Wollscheid B. Mapping the dynamic high-density lipoprotein synapse. Atherosclerosis 2023; 380:117200. [PMID: 37619408 DOI: 10.1016/j.atherosclerosis.2023.117200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND AND AIMS Heterogeneous high-density lipoprotein (HDL) particles, which can contain hundreds of proteins, affect human health and disease through dynamic molecular interactions with cell surface proteins. How HDL mediates its long-range signaling functions and interactions with various cell types is largely unknown. Due to the complexity of HDL, we hypothesize that multiple receptors engage with HDL particles resulting in condition-dependent receptor-HDL interaction clusters at the cell surface. METHODS Here we used the mass spectrometry-based and light-controlled proximity labeling strategy LUX-MS in a discovery-driven manner to decode HDL-receptor interactions. RESULTS Surfaceome nanoscale organization analysis of hepatocytes and endothelial cells using LUX-MS revealed that the previously known HDL-binding protein scavenger receptor B1 (SCRB1) is embedded in a cell surface protein community, which we term HDL synapse. Modulating the endothelial HDL synapse, composed of 60 proteins, by silencing individual members, showed that the HDL synapse can be assembled in the absence of SCRB1 and that the members are interlinked. The aminopeptidase N (AMPN) (also known as CD13) was identified as an HDL synapse member that directly influences HDL uptake into the primary human aortic endothelial cells (HAECs). CONCLUSIONS Our data indicate that preformed cell surface residing protein complexes modulate HDL function and suggest new theragnostic opportunities.
Collapse
Affiliation(s)
- Kathrin Frey
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland.
| | - Lucia Rohrer
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Fabian Frommelt
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Meret Ringwald
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Anton Potapenko
- Institute for Clinical Chemistry University Hospital Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland
| | | | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland; ETH PHRT Swiss Multi-Omics Center (SMOC), Switzerland.
| |
Collapse
|
34
|
Ledesma-Durán A, Juárez-Valencia LH. Diffusion coefficients and MSD measurements on curved membranes and porous media. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:70. [PMID: 37578670 DOI: 10.1140/epje/s10189-023-00329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/28/2023] [Indexed: 08/15/2023]
Abstract
We study some geometric aspects that influence the transport properties of particles that diffuse on curved surfaces. We compare different approaches to surface diffusion based on the Laplace-Beltrami operator adapted to predict concentration along entire membranes, confined subdomains along surfaces, or within porous media. Our goal is to summarize, firstly, how diffusion in these systems results in different types of diffusion coefficients and mean square displacement measurements, and secondly, how these two factors are affected by the concavity of the surface, the shape of the possible barriers or obstacles that form the available domains, the sinuosity, tortuosity, and constrictions of the trajectories and even how the observation plane affects the measurements of the diffusion. In addition to presenting a critical and organized comparison between different notions of MSD, in this review, we test the correspondence between theoretical predictions and numerical simulations by performing finite element simulations and illustrate some situations where diffusion theory can be applied. We briefly reviewed computational schemes for understanding surface diffusion and finally, discussed how this work contributes to understanding the role of surface diffusion transport properties in porous media and their relationship to other transport processes.
Collapse
Affiliation(s)
- Aldo Ledesma-Durán
- Departmento de Matemáticas, Universidad Autónoma Metropolitana, CDMX, Mexico
| | | |
Collapse
|
35
|
Huang Z, Kanchanawong P. Ultra high-speed single-molecule fluorescence imaging. J Cell Biol 2023; 222:e202306136. [PMID: 37458726 PMCID: PMC10351246 DOI: 10.1083/jcb.202306136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
In two articles in this issue, Fujiwara et al. developed an ultrasensitive high-speed camera capable of single-molecule fluorescence imaging at a microsecond timescale (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202110160). This major leap in detection speed enables the organization of plasma membrane and integrin-based adhesions to be probed in unprecedented detail (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202110162).
Collapse
Affiliation(s)
- Zengxin Huang
- Mechanobiology Institute, National University of Singapore, Singapore, Republic of Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Republic of Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
36
|
Wu J, Xu C, Ye Z, Chen H, Wang Y, Yang K, Yuan B. Transition between Different Diffusion Modes of Individual Lipids during the Membrane-Specific Action of As-CATH4 Peptides. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301713. [PMID: 37093200 DOI: 10.1002/smll.202301713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/02/2023] [Indexed: 05/03/2023]
Abstract
The cell membrane permeabilization ability of immune defense antimicrobial peptides (AMPs) is widely applied in biomedicine. Although the mechanisms of peptide-membrane interactions have been widely investigated, analyses at the molecular level are still lacking. Herein, the membrane-specific action of a native AMP, As-CATH4, is investigated using a single-lipid tracking method in combination with live cell and model membrane assays conducted at different scales. The peptide-membrane interaction process is characterized by analyzing single-lipid diffusion behaviors. As-CATH4 exhibits potent antimicrobial activity through bacterial membrane permeabilization, with moderate cytotoxicity against mammalian cells. In-plane diffusion analyses of individual lipids show that the lipid molecules exhibit non-Gaussian and heterogeneous diffusion behaviors in both pristine and peptide-treated membranes, which can be decomposed into two Gaussian subgroups corresponding to normal- and slow-diffusive lipids. Assessment of the temporal evolution of these two diffusion modes of lipids reveal that the peptide action states of As-CATH4 include surface binding, transmembrane defect formation, and dynamic equilibrium. The action mechanisms of As-CATH4 at varying concentrations and against different membranes are distinguished. This work resolves the simultaneous mixed diffusion mechanisms of single lipids in biomimetic cell membranes, especially during dynamic membrane permeabilization by AMPs.
Collapse
Affiliation(s)
- Jinfeng Wu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, 215006, P. R. China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, P. R. China
| | - Cheng Xu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, 215006, P. R. China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, P. R. China
| | - Zifan Ye
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Haibo Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, 215006, P. R. China
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, 215006, P. R. China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, P. R. China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, P. R. China
| |
Collapse
|
37
|
Thallmair V, Schultz L, Evers S, Jolie T, Goecke C, Leitner MG, Thallmair S, Oliver D. Localization of the tubby domain, a PI(4,5)P2 biosensor, to E-Syt3-rich endoplasmic reticulum-plasma membrane junctions. J Cell Sci 2023; 136:jcs260848. [PMID: 37401342 PMCID: PMC10445746 DOI: 10.1242/jcs.260848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/26/2023] [Indexed: 07/05/2023] Open
Abstract
The phospholipid phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2] acts as a signaling lipid at the plasma membrane (PM) with pleiotropic regulatory actions on multiple cellular processes. Signaling specificity might result from spatiotemporal compartmentalization of the lipid and from combinatorial binding of PI(4,5)P2 effector proteins to additional membrane components. Here, we analyzed the spatial distribution of tubbyCT, a paradigmatic PI(4,5)P2-binding domain, in live mammalian cells by total internal reflection fluorescence (TIRF) microscopy and molecular dynamics simulations. We found that unlike other well-characterized PI(4,5)P2 recognition domains, tubbyCT segregates into distinct domains within the PM. TubbyCT enrichment occurred at contact sites between PM and endoplasmic reticulum (ER) (i.e. at ER-PM junctions) as shown by colocalization with ER-PM markers. Localization to these sites was mediated in a combinatorial manner by binding to PI(4,5)P2 and by interaction with a cytosolic domain of extended synaptotagmin 3 (E-Syt3), but not other E-Syt isoforms. Selective localization to these structures suggests that tubbyCT is a novel selective reporter for a ER-PM junctional pool of PI(4,5)P2. Finally, we found that association with ER-PM junctions is a conserved feature of tubby-like proteins (TULPs), suggesting an as-yet-unknown function of TULPs.
Collapse
Affiliation(s)
- Veronika Thallmair
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps UniversityMarburg, 35037 Marburg, Germany
| | - Lea Schultz
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Saskia Evers
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Theresa Jolie
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Christian Goecke
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
| | - Michael G. Leitner
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH&Co.KG, Birkendorfer Str. 65, 88400 Biberach an der Riß, Germany
| | - Sebastian Thallmair
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt am Main, Germany
- Groningen Biomolecular Sciences and Biotechnology Institute and The Zernike Institute for Advanced Material, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, 35037 Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps UniversityMarburg, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, 35032 Marburg, Germany
| |
Collapse
|
38
|
Henning P, Köster T, Haack F, Burrage K, Uhrmacher AM. Implications of different membrane compartmentalization models in particle-based in silico studies. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221177. [PMID: 37416823 PMCID: PMC10320350 DOI: 10.1098/rsos.221177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
Studying membrane dynamics is important to understand the cellular response to environmental stimuli. A decisive spatial characteristic of the plasma membrane is its compartmental structure created by the actin-based membrane-skeleton (fences) and anchored transmembrane proteins (pickets). Particle-based reaction-diffusion simulation of the membrane offers a suitable temporal and spatial resolution to analyse its spatially heterogeneous and stochastic dynamics. Fences have been modelled via hop probabilities, potentials or explicit picket fences. Our study analyses the different approaches' constraints and their impact on simulation results and performance. Each of the methods comes with its own constraints; the picket fences require small timesteps, potential fences might induce a bias in diffusion in crowded systems, and probabilistic fences, in addition to carefully scaling the probability with the timesteps, induce higher computational costs for each propagation step.
Collapse
Affiliation(s)
- Philipp Henning
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Till Köster
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Fiete Haack
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Kevin Burrage
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
- Visiting Professor, Department of Computer Science, University of Oxford, Oxford, UK
| | - Adelinde M. Uhrmacher
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| |
Collapse
|
39
|
Zakany F, Mándity IM, Varga Z, Panyi G, Nagy P, Kovacs T. Effect of the Lipid Landscape on the Efficacy of Cell-Penetrating Peptides. Cells 2023; 12:1700. [PMID: 37443733 PMCID: PMC10340183 DOI: 10.3390/cells12131700] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Every cell biological textbook teaches us that the main role of the plasma membrane is to separate cells from their neighborhood to allow for a controlled composition of the intracellular space. The mostly hydrophobic nature of the cell membrane presents an impenetrable barrier for most hydrophilic molecules larger than 1 kDa. On the other hand, cell-penetrating peptides (CPPs) are capable of traversing this barrier without compromising membrane integrity, and they can do so on their own or coupled to cargos. Coupling biologically and medically relevant cargos to CPPs holds great promise of delivering membrane-impermeable drugs into cells. If the cargo is able to interact with certain cell types, uptake of the CPP-drug complex can be tailored to be cell-type-specific. Besides outlining the major membrane penetration pathways of CPPs, this review is aimed at deciphering how properties of the membrane influence the uptake mechanisms of CPPs. By summarizing an extensive body of experimental evidence, we argue that a more ordered, less flexible membrane structure, often present in the very diseases planned to be treated with CPPs, decreases their cellular uptake. These correlations are not only relevant for understanding the cellular biology of CPPs, but also for rationally improving their value in translational or clinical applications.
Collapse
Affiliation(s)
- Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (F.Z.); (Z.V.); (G.P.)
| | - István M. Mándity
- Department of Organic Chemistry, Faculty of Pharmacy, Semmelweis University, 1085 Budapest, Hungary;
- TTK Lendület Artificial Transporter Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (F.Z.); (Z.V.); (G.P.)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (F.Z.); (Z.V.); (G.P.)
| | - Peter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (F.Z.); (Z.V.); (G.P.)
| | - Tamas Kovacs
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (F.Z.); (Z.V.); (G.P.)
| |
Collapse
|
40
|
Müller GA, Müller TD. (Patho)Physiology of Glycosylphosphatidylinositol-Anchored Proteins II: Intercellular Transfer of Matter (Inheritance?) That Matters. Biomolecules 2023; 13:994. [PMID: 37371574 DOI: 10.3390/biom13060994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (APs) are anchored at the outer leaflet of the plasma membrane (PM) bilayer by covalent linkage to a typical glycolipid and expressed in all eukaryotic organisms so far studied. Lipolytic release from PMs into extracellular compartments and intercellular transfer are regarded as the main (patho)physiological roles exerted by GPI-APs. The intercellular transfer of GPI-APs relies on the complete GPI anchor and is mediated by extracellular vesicles such as microvesicles and exosomes and lipid-free homo- or heteromeric aggregates, and lipoprotein-like particles such as prostasomes and surfactant-like particles, or lipid-containing micelle-like complexes. In mammalian organisms, non-vesicular transfer is controlled by the distance between donor and acceptor cells/tissues; intrinsic conditions such as age, metabolic state, and stress; extrinsic factors such as GPI-binding proteins; hormones such as insulin; and drugs such as anti-diabetic sulfonylureas. It proceeds either "directly" upon close neighborhood or contact of donor and acceptor cells or "indirectly" as a consequence of the induced lipolytic release of GPI-APs from PMs. Those displace from the serum GPI-binding proteins GPI-APs, which have retained the complete anchor, and become assembled in aggregates or micelle-like complexes. Importantly, intercellular transfer of GPI-APs has been shown to induce specific phenotypes such as stimulation of lipid and glycogen synthesis, in cultured human adipocytes, blood cells, and induced pluripotent stem cells. As a consequence, intercellular transfer of GPI-APs should be regarded as non-genetic inheritance of (acquired) features between somatic cells which is based on the biogenesis and transmission of matter such as GPI-APs and "membrane landscapes", rather than the replication and transmission of information such as DNA. Its operation in mammalian organisms remains to be clarified.
Collapse
Affiliation(s)
- Günter A Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD) at the Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) and German Center for Diabetes Research (DZD) at the Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
41
|
Liu M, van Kuppeveld FJM, de Haan CAM, de Vries E. Gradual adaptation of animal influenza A viruses to human-type sialic acid receptors. Curr Opin Virol 2023; 60:101314. [DOI: 10.1016/j.coviro.2023.101314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
|
42
|
Arnold DP, Xu Y, Takatori SC. Antibody binding reports spatial heterogeneities in cell membrane organization. Nat Commun 2023; 14:2884. [PMID: 37208326 DOI: 10.1038/s41467-023-38525-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
The spatial organization of cell membrane glycoproteins and glycolipids is critical for mediating the binding of ligands, receptors, and macromolecules on the plasma membrane. However, we currently do not have the methods to quantify the spatial heterogeneities of macromolecular crowding on live cell surfaces. In this work, we combine experiment and simulation to report crowding heterogeneities on reconstituted membranes and live cell membranes with nanometer spatial resolution. By quantifying the effective binding affinity of IgG monoclonal antibodies to engineered antigen sensors, we discover sharp gradients in crowding within a few nanometers of the crowded membrane surface. Our measurements on human cancer cells support the hypothesis that raft-like membrane domains exclude bulky membrane proteins and glycoproteins. Our facile and high-throughput method to quantify spatial crowding heterogeneities on live cell membranes may facilitate monoclonal antibody design and provide a mechanistic understanding of plasma membrane biophysical organization.
Collapse
Affiliation(s)
- Daniel P Arnold
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Yaxin Xu
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Sho C Takatori
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
43
|
Nguyen N, Lewis A, Pham T, Sikazwe D, Cheng KH. Exploring the Role of Anionic Lipid Nanodomains in the Membrane Disruption and Protein Folding of Human Islet Amyloid Polypeptide Oligomers on Lipid Membrane Surfaces Using Multiscale Molecular Dynamics Simulations. Molecules 2023; 28:4191. [PMID: 37241931 PMCID: PMC10223233 DOI: 10.3390/molecules28104191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The aggregation of human Islet Amyloid Polypeptide (hIAPP) on cell membranes is linked to amyloid diseases. However, the physio-chemical mechanisms of how these hIAPP aggregates trigger membrane damage are unclear. Using coarse-grained and all-atom molecular dynamics simulations, we investigated the role of lipid nanodomains in the presence or absence of anionic lipids, phosphatidylserine (PS), and a ganglioside (GM1), in the membrane disruption and protein folding behaviors of hIAPP aggregates on phase-separated raft membranes. Our raft membranes contain liquid-ordered (Lo), liquid-disordered (Ld), mixed Lo/Ld (Lod), PS-cluster, and GM1-cluster nanosized domains. We observed that hIAPP aggregates bound to the Lod domain in the absence of anionic lipids, but also to the GM1-cluster- and PS-cluster-containing domains, with stronger affinity in the presence of anionic lipids. We discovered that L16 and I26 are the lipid anchoring residues of hIAPP binding to the Lod and PS-cluster domains. Finally, significant lipid acyl chain order disruption in the annular lipid shells surrounding the membrane-bound hIAPP aggregates and protein folding, particularly beta-sheet formation, in larger protein aggregates were evident. We propose that the interactions of hIAPP and both non-anionic and anionic lipid nanodomains represent key molecular events of membrane damage associated with the pathogenesis of amyloid diseases.
Collapse
Affiliation(s)
- Ngoc Nguyen
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
| | - Amber Lewis
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA;
| | - Thuong Pham
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
| | - Donald Sikazwe
- Pharmaceutical Sciences Department, Feik School of Pharmacy, University of the Incarnate Word, San Antonio, TX 78209, USA;
| | - Kwan H. Cheng
- Physics Department, Trinity University, San Antonio, TX 78212, USA; (N.N.); (T.P.)
- Neuroscience Department, Trinity University, San Antonio, TX 78212, USA;
| |
Collapse
|
44
|
Kusumi A, Tsunoyama TA, Tang B, Hirosawa KM, Morone N, Fujiwara TK, Suzuki KGN. Cholesterol- and actin-centered view of the plasma membrane: updating the Singer-Nicolson fluid mosaic model to commemorate its 50th anniversary †. Mol Biol Cell 2023; 34:pl1. [PMID: 37039596 PMCID: PMC10162409 DOI: 10.1091/mbc.e20-12-0809] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/07/2022] [Accepted: 02/07/2023] [Indexed: 04/12/2023] Open
Abstract
Two very polarized views exist for understanding the cellular plasma membrane (PM). For some, it is the simple fluid described by the original Singer-Nicolson fluid mosaic model. For others, due to the presence of thousands of molecular species that extensively interact with each other, the PM forms various clusters and domains that are constantly changing and therefore, no simple rules exist that can explain the structure and molecular dynamics of the PM. In this article, we propose that viewing the PM from its two predominant components, cholesterol and actin filaments, provides an excellent and transparent perspective of PM organization, dynamics, and mechanisms for its functions. We focus on the actin-induced membrane compartmentalization and lipid raft domains coexisting in the PM and how they interact with each other to perform PM functions. This view provides an important update of the fluid mosaic model.
Collapse
Affiliation(s)
- Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Taka A. Tsunoyama
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
| | - Bo Tang
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904-0495, Japan
| | - Koichiro M. Hirosawa
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Nobuhiro Morone
- MRC Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK
| | - Takahiro K. Fujiwara
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Kenichi G. N. Suzuki
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
45
|
Nicolson GL, Ferreira de Mattos G. The Fluid-Mosaic model of cell membranes: A brief introduction, historical features, some general principles, and its adaptation to current information. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184135. [PMID: 36746313 DOI: 10.1016/j.bbamem.2023.184135] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
The Fluid-Mosaic Membrane (FMM) model was originally proposed as a general, nanometer-scale representation of cell membranes (Singer and Nicolson, 1972). The FMM model was based on some general principles, such as thermodynamic considerations, intercalation of globular proteins into a lipid bilayer, independent protein and lipid dynamics, cooperativity and other characteristics. Other models had trimolecular structures or membrane globular lipoprotein units. These latter models were flawed, because they did not allow autonomous lipids, membrane domains or discrete lateral dynamics. The FMM model was also consistent with membrane asymmetry, cis- and trans-membrane linkages and associations of membrane components into multi-molecular complexes and domains. It has remained useful for explaining the basic organizational principles and properties of various biological membranes. New information has been added, such as membrane-associated cytoskeletal assemblies, extracellular matrix interactions, transmembrane controls, specialized lipid-protein domains that differ in compositions, rotational and lateral mobilities, lifetimes, functions, and other characteristics. The presence of dense, structured membrane domains has reduced significantly the extent of fluid-lipid membrane areas, and the FMM model is now considered to be more mosaic and dense than the original proposal.
Collapse
Affiliation(s)
- Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA.
| | - Gonzalo Ferreira de Mattos
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
46
|
Tuomivaara ST, Teo CF, Jan YN, Jan LY, Wiita AP. SLAPSHOT reveals rapid dynamics of extracellularly exposed proteome in response to calcium-activated plasma membrane phospholipid scrambling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.26.534250. [PMID: 36993417 PMCID: PMC10055316 DOI: 10.1101/2023.03.26.534250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
To facilitate our understanding of the often rapid and nuanced dynamics of extracellularly exposed proteomes during signaling events, it is important to devise robust workflows affording fast time resolution without biases and confounding factors. Here, we present Surface-exposed protein Labeling using PeroxidaSe, H2O2, and Tyramide-derivative (SLAPSHOT), to label extracellularly exposed proteins in a rapid, sensitive, and specific manner, while preserving cellular integrity. This experimentally simple and flexible method utilizes recombinant soluble APEX2 peroxidase that is applied to cells, thus circumventing biological perturbations, tedious engineering of tools and cells, and labeling biases. APEX2 neither requires metal cations for activity nor contains disulfide bonds, conferring versatility for a wide spectrum of experimental setups. We applied SLAPSHOT followed by quantitative mass spectrometry-based proteomics analysis to examine the immediate and extensive cell surface expansion and ensuing restorative membrane shedding upon the activation of Scott syndrome-linked TMEM16F, a ubiquitously expressed calcium-dependent phospholipid scramblase and ion channel. Time-course data ranging from one to thirty minutes of calcium stimulation using wild-type and TMEM16F deficient cells revealed intricate co-regulation of known protein families, including those in the integrin and ICAM families. Crucially, we identified proteins that are known to reside in intracellular organelles, including ER, as occupants of the freshly deposited membrane, and mitovesicles as an abundant component and contributor to the extracellularly exposed proteome. Our study not only provides the first accounts of the immediate consequences of calcium signaling on the extracellularly exposed proteome, but also presents a blueprint for the application of SLAPSHOT as a general approach for monitoring extracellularly exposed protein dynamics.
Collapse
Affiliation(s)
- Sami T. Tuomivaara
- Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Chin Fen Teo
- Howard Hughes Medical Institute, University of California, San Francisco, CA
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, University of California, San Francisco, CA
- Department of Physiology, University of California, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA
| | - Lily Y. Jan
- Howard Hughes Medical Institute, University of California, San Francisco, CA
- Department of Physiology, University of California, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA
| | - Arun P. Wiita
- Department of Laboratory Medicine, University of California, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
47
|
Ullo MF, Case LB. How cells sense and integrate information from different sources. WIREs Mech Dis 2023:e1604. [PMID: 36781396 DOI: 10.1002/wsbm.1604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/06/2023] [Accepted: 01/24/2023] [Indexed: 02/15/2023]
Abstract
Cell signaling is a fundamental cellular process that enables cells to sense and respond to information in their surroundings. At the molecular level, signaling is primarily carried out by transmembrane protein receptors that can initiate complex downstream signal transduction cascades to alter cellular behavior. In the human body, different cells can be exposed to a wide variety of environmental conditions, and cells express diverse classes of receptors capable of sensing and integrating different signals. Furthermore, different receptors and signaling pathways can crosstalk with each other to calibrate the cellular response. Crosstalk occurs through multiple mechanisms at different levels of signaling pathways. In this review, we discuss how cells sense and integrate different chemical, mechanical, and spatial signals as well as the mechanisms of crosstalk between pathways. To illustrate these concepts, we use a few well-studied signaling pathways, including receptor tyrosine kinases and integrin receptors. Finally, we discuss the implications of dysregulated cellular sensing on driving diseases such as cancer. This article is categorized under: Cancer > Molecular and Cellular Physiology Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Maria F Ullo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lindsay B Case
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
48
|
Refinement of Singer-Nicolson fluid-mosaic model by microscopy imaging: Lipid rafts and actin-induced membrane compartmentalization. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184093. [PMID: 36423676 DOI: 10.1016/j.bbamem.2022.184093] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
This year celebrates the 50th anniversary of the Singer-Nicolson fluid mosaic model for biological membranes. The next level of sophistication we have achieved for understanding plasma membrane (PM) structures, dynamics, and functions during these 50 years includes the PM interactions with cortical actin filaments and the partial demixing of membrane constituent molecules in the PM, particularly raft domains. Here, first, we summarize our current knowledge of these two structures and emphasize that they are interrelated. Second, we review the structure, molecular dynamics, and function of raft domains, with main focuses on raftophilic glycosylphosphatidylinositol-anchored proteins (GPI-APs) and their signal transduction mechanisms. We pay special attention to the results obtained by single-molecule imaging techniques and other advanced microscopy methods. We also clarify the limitations of present optical microscopy methods for visualizing raft domains, but emphasize that single-molecule imaging techniques can "detect" raft domains associated with molecules of interest in the PM.
Collapse
|
49
|
Dasgupta A, Ngo HT, Tschoerner D, Touret N, da Rocha-Azevedo B, Jaqaman K. Multiscale imaging and quantitative analysis of plasma membrane protein-cortical actin interplay. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525112. [PMID: 36747866 PMCID: PMC9900770 DOI: 10.1101/2023.01.22.525112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The spatiotemporal organization of cell surface receptors is important for cell signaling. Cortical actin (CA), the subset of the actin cytoskeleton subjacent to the plasma membrane (PM), plays a large role in cell surface receptor organization. This was however shown largely through actin perturbation experiments, which raise concerns of nonspecific effects and preclude quantification of actin architecture and dynamics under unperturbed conditions. These limitations make it challenging to predict how changes in CA properties can affect receptor organization. To derive direct relationships between the architecture and dynamics of CA and the spatiotemporal organization of PM proteins, including cell surface receptors, we developed a multiscale imaging and computational analysis framework based on the integration of single-molecule imaging (SMI) of PM proteins and fluorescent speckle microscopy (FSM) of CA (combined: SMI-FSM) in the same live cell. SMI-FSM revealed differential relationships between PM proteins and CA based on the PM proteins’ actin binding ability, diffusion type and local CA density. It also highlighted the complexity of cell wide actin perturbation, where we found that global changes in actin properties caused by perturbation were not necessarily reflected in the CA properties near PM proteins, and the changes in PM protein properties upon perturbation varied based on the local CA environment. Given the widespread use of SMI as a method to study the spatiotemporal organization of PM proteins and the versatility of SMI-FSM, we expect it to be widely applicable to enable future investigation of the influence of CA architecture and dynamics on different PM proteins, especially in the context of actin-dependent cellular processes, such as cell migration. Significance Plasma membrane protein organization, an important factor for shaping cellular behaviors, is influenced by cortical actin, the subset of the actin cytoskeleton near the plasma membrane. Yet it is challenging to directly and quantitatively probe this influence. Here, we developed an imaging and analysis approach that combines single-molecule imaging, fluorescent speckle microscopy and computational statistical analysis to characterize and correlate the spatiotemporal organization of plasma membrane proteins and cortical actin. Our approach revealed different relationships between different proteins and cortical actin, and highlighted the complexity of interpreting cell wide actin perturbation experiments. We expect this approach to be widely used to study the influence of cortical actin on different plasma membrane components, especially in actin-dependent processes.
Collapse
Affiliation(s)
- Aparajita Dasgupta
- Department of Biophysics, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Huong-Tra Ngo
- Department of Biophysics, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Deryl Tschoerner
- Department of Biophysics, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Nicolas Touret
- Department of Biochemistry, University of Alberta; Edmonton, AB, Canada
| | - Bruno da Rocha-Azevedo
- Department of Biophysics, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Khuloud Jaqaman
- Department of Biophysics, University of Texas Southwestern Medical Center; Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center; Dallas, TX, USA
| |
Collapse
|
50
|
Andersen C, Zulueta Díaz YDLM, Kure JL, Hessellund Eriksen M, Lovatt AL, Lagerholm C, Morales S, Sehayek S, Sheard TMD, Wiseman PW, Arnspang EC. Angiotensin II Treatment Induces Reorganization and Changes in the Lateral Dynamics of Angiotensin II Type 1 Receptor in the Plasma Membrane Elucidated by Photoactivated Localization Microscopy Combined with Image Spatial Correlation Analysis. Anal Chem 2023; 95:730-738. [PMID: 36574961 DOI: 10.1021/acs.analchem.2c02720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mechanisms by which angiotensin II type 1 receptor is distributed and the diffusional pattern in the plasma membrane (PM) remain unclear, despite their crucial role in cardiovascular homeostasis. In this work, we obtained quantitative information of angiotensin II type 1 receptor (AT1R) lateral dynamics as well as changes in the diffusion properties after stimulation with ligands in living cells using photoactivated localization microscopy (PALM) combined with image spatial-temporal correlation analysis. To study the organization of the receptor at the nanoscale, expansion microscopy (ExM) combined with PALM was performed. This study revealed that AT1R lateral diffusion increased after binding to angiotensin II (Ang II) and the receptor diffusion was transiently confined in the PM. In addition, ExM revealed that AT1R formed nanoclusters at the PM and the cluster size significantly decreased after Ang II treatment. Taking these results together suggest that Ang II binding and activation cause reorganization and changes in the dynamics of AT1R at the PM.
Collapse
Affiliation(s)
- Camilla Andersen
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Odense M5230, Denmark
| | | | - Jakob L Kure
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Odense M5230, Denmark
| | - Mathias Hessellund Eriksen
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Odense M5230, Denmark
| | - Adam Leslie Lovatt
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Odense M5230, Denmark
| | | | - Sebastian Morales
- Department of Physics and Department of Chemistry, McGill University, MontrealH3A 0B8, Canada
| | - Simon Sehayek
- Department of Physics and Department of Chemistry, McGill University, MontrealH3A 0B8, Canada
| | - Thomas M D Sheard
- School of Biosciences, University of Sheffield, SheffieldS10 2TN, U.K
| | - Paul W Wiseman
- Department of Physics and Department of Chemistry, McGill University, MontrealH3A 0B8, Canada
| | - Eva C Arnspang
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Odense M5230, Denmark
| |
Collapse
|