1
|
Williams JK, Ngo JM, Murugupandiyan A, Croall DE, Hartzell HC, Schekman R. Calpains orchestrate secretion of annexin-containing microvesicles during membrane repair. J Cell Biol 2025; 224:e202408159. [PMID: 40377476 PMCID: PMC12083247 DOI: 10.1083/jcb.202408159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/04/2025] [Accepted: 04/02/2025] [Indexed: 05/18/2025] Open
Abstract
Microvesicles (MVs) are membrane-enclosed, plasma membrane-derived particles released by cells from all branches of life. MVs have utility as disease biomarkers and may participate in intercellular communication; however, physiological processes that induce their secretion are not known. Here, we isolate and characterize annexin-containing MVs and show that these vesicles are secreted in response to the calcium influx caused by membrane damage. The annexins in these vesicles are cleaved by calpains. After plasma membrane injury, cytoplasmic calcium-bound annexins are rapidly recruited to the plasma membrane and form a scab-like structure at the lesion. In a second phase, recruited annexins are cleaved by calpains-1/2, disabling membrane scabbing. Cleavage promotes annexin secretion within MVs. Our data support a new model of plasma membrane repair, where calpains relax annexin-membrane aggregates in the lesion repair scab, allowing secretion of damaged membrane and annexins as MVs. We anticipate that cells experiencing plasma membrane damage, including muscle and metastatic cancer cells, secrete these MVs at elevated levels.
Collapse
Affiliation(s)
- Justin Krish Williams
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jordan Matthew Ngo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | | - Dorothy E. Croall
- Department of Biochemistry, Microbiology and Molecular Biology, University of Maine, Orono, ME, USA
| | - H. Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
2
|
Nakao H, Tsujii T, Saito H, Ikeda K, Nakano M. Synergistic effects of hydrophilic residues in the transmembrane region on lipid scrambling activity of dimeric helices. Colloids Surf B Biointerfaces 2025; 251:114612. [PMID: 40086211 DOI: 10.1016/j.colsurfb.2025.114612] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Phospholipid scramblases promote lipid transbilayer movement (flip-flop) in the plasma membrane, which is involved in a wide range of cellular functions, such as phagocytosis and blood coagulation. One structural characteristic of scramblases and model lipid scrambling peptides is the presence of hydrophilic residues in their transmembrane domains. These hydrophilic regions are considered the active sites through which lipid polar headgroups pass during the translocation process. However, how the structural arrangement of hydrophilic residues results in strong lipid scrambling activities in scramblases needs to be investigated, because the effects of a single hydrophilic residue on lipid scrambling are much lower than the activity of natural scramblases. Here, we developed double-spanning transmembrane peptides containing varying numbers of Gln residues. A combination of lipid vesicle experiments and molecular dynamics simulations indicates that lipid scrambling activities are synergistically enhanced by the proximity between planes created by Gln residues aligned parallel to the helix and that interactions between Gln and Trp residues stabilize the strongly active structures. The contribution of Gln residues to lipid scrambling activity suggests that the alignment and proximity of hydrophilic residues in the transmembrane region is one of the mechanisms of lipid scrambling by natural scramblases. This study provides clues for the energetic and structural mechanisms of lipid scrambling and for the design of artificial phospholipid scramblases.
Collapse
Affiliation(s)
- Hiroyuki Nakao
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Toshiki Tsujii
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Hiroaki Saito
- Faculty of Pharmaceutical Sciences, Hokuriku University, 3 Kanagawamachi, Kanazawa, Ishikawa 920-1181, Japan
| | - Keisuke Ikeda
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Minoru Nakano
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
3
|
Wu Y, Liu Q, Tian R, Zhao S, Ding B, Jiang Q. Predesigned DNA Origami Nanodrills Mediate Controlled Pore Formation on a Plasma Membrane and Cell Death. NANO LETTERS 2025; 25:8220-8226. [PMID: 40343847 DOI: 10.1021/acs.nanolett.5c01290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
We presented an engineered DNA origami nanodrill that is capable of controlled attachment and penetration of the plasma membrane, thereby inducing cell damage and lytic death. The cap-and-stem-like DNA origami nanodrills were constructed, equipping them with addressable cholesterol tags on their caps for adherence to lipid bilayers. The subsequent insertion of the origami stems was initiated, enabling the generation of distinct transmembrane channels in various artificial and biological membranes. These nanodrills with plasma-membrane-disrupting functions resulted in mechanical damage to living cell membranes, leading to increased cytosolic calcium levels, enhanced intracellular protein release, and potent cytotoxicity against tumor cells. The further stimuli-responsive design enabled acid-triggered attachment of DNA nanodrills to the plasma membrane and subsequent cell damage in a pH-controlled fashion. This programmable origami nanodrill could serve as an artificial mediator of plasma-membrane rupture and cell death, thus potentially leading to the development of new therapeutic strategies to combat cancer.
Collapse
Affiliation(s)
- Yushuai Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Run Tian
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Zhao
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Sputay D, Doktorova M, Chan SH, Guo EH, Wang HY, Lorent JH, Levental I, Levental KR. Immune cell activation produces locally scrambled foci of plasma membrane lipids. Faraday Discuss 2025. [PMID: 40351229 PMCID: PMC12067185 DOI: 10.1039/d4fd00205a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/11/2025] [Indexed: 05/14/2025]
Abstract
Most eukaryotic cells maintain a large disparity in lipid compositions between the cytosolic and external leaflets of the plasma membrane (PM) bilayer. This lipid asymmetry is maintained by energy-consuming flippase enzymes that selectively drive phospholipids into the cytosolic leaflet, often against large concentration gradients. Scramblases, activated by intracellular Ca2+ or apoptotic signaling, shuttle phospholipids down their concentration gradient to release lipid asymmetry. Such scrambling is typically evidenced by exposure of phosphatidylserine (PS) to the external leaflet and is associated with many physiological processes, most notably blood clotting and cell death, but also activation of immune cells. Here, we show that both PS and phosphatidylethanolamine (PE) appear on the PM external leaflet following immune receptor-mediated activation of mast cells. We also observe similar effects in T cells. Importantly, in contrast to wholesale release of PM asymmetry induced by calcium ionophores or apoptosis, we show that scrambling in activated immune cells is focal, with small, stable regions of surface exposed PS. These scrambled foci are calcium dependent, have lower lipid packing than their surrounding outer leaflet, and are reversible. These observations of local, transient scrambling during physiological activation of healthy immune cells suggest important roles for the lateral and transbilayer organization of membrane lipids.
Collapse
Affiliation(s)
- Daryna Sputay
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Milka Doktorova
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Sweden
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Emma Han Guo
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Joseph H Lorent
- Cellular and Molecular Pharmacology (FACM), Louvain Drug Research Institute, UCLouvain, Belgium
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA.
| |
Collapse
|
5
|
Gadiyar V, Pulica R, Aquib A, Tranos JA, Varsanyi C, Almansa LF, Gaspers L, Lorenzo MD, Kotenko SV, Tripathi S, Howell RW, Choudhary A, Calianese DC, Birge RB. Phospholipid Scramblases TMEM16F and Xkr8 regulate distinct features of Phosphatidylserine (PS) externalization and immune regulation in the tumor microenvironment to regulate tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.17.649445. [PMID: 40391322 PMCID: PMC12087989 DOI: 10.1101/2025.04.17.649445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The phospholipid scramblases Xkr8 and TMEM16F externalize phosphatidylserine (PS) on cells by distinct molecular mechanisms. Xkr8, a caspase-activated scramblase, is activated by caspase-mediated proteolytic cleavage, and in synergy with caspase-mediated inactivation of P4-type ATP-dependent flippases, results in the irreversible externalization of PS on the dying cells and an "eat-me" signal for efferocytosis. In contrast, TMEM16F is a calcium activated scramblase that reversibly externalizes PS on viable cells via the transient increase in intracellular calcium on activated or growth factor stimulated cells. By contrast to the abovementioned homeostatic mechanisms of PS externalization under physiological conditions, PS becomes constitutively externalized in the tumor microenvironment (TME) in many solid tumor types by a complex mechanistic, posited both via the high apoptotic indexes of tumors, but also by the prolonged oncogenic and metabolic stresses that occur in the TME. Such chronic and persistent PS externalization in the TME has been linked to host immune evasion and the tonic interactions of PS with inhibitory PS receptors such as TAM (Tyro3, Axl, Mertk) and TIM (T cell/transmembrane, immunoglobulin, and mucin) family receptors. Here, in an effort to better understand the contributions of apoptotic vs live cell PS-externalization with respect to tumorigenesis and immune evasion, we employed an E0771 luminal B breast cancer orthotopic in vivo model and genetically ablated Xkr8 and TMEM16F using CRISPR/Cas9. While neither the knockout of Xkr8 nor TMEM16F showed defects in cell intrinsic properties related to cell growth, tumor sphere formation, cell migration, and growth factor signaling, both knockouts suppressed tumorigenicity in immune-competent mice, but not in NOD/SCID or RAG deficient immune-deficient strains. Mechanistically, at the cell biological level, Xkr8 knockout suppressed macrophage-mediated efferocytosis, and TMEM16F knockout suppressed ER stress/calcium-induced PS externalization. Our data support an emerging idea in immune-oncology and immunotherapy that constitutive PS externalization, mediated by the activation of scramblases on tumor cells, can support immune evasion in the tumor microenvironment thereby linking a combination of apoptosis/efferocytosis and oncogenic stress involving calcium dysregulation the contribute to PS-mediated immune escape and cancer progression.
Collapse
|
6
|
Pulica R, Aquib A, Varsanyi C, Gadiyar V, Wang Z, Frederick T, Calianese DC, Patel B, de Dios KV, Poalasin V, De Lorenzo MS, Kotenko SV, Wu Y, Yang A, Choudhary A, Sriram G, Birge RB. Dys-regulated phosphatidylserine externalization as a cell intrinsic immune escape mechanism in cancer. Cell Commun Signal 2025; 23:131. [PMID: 40069722 PMCID: PMC11900106 DOI: 10.1186/s12964-025-02090-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
The negatively charged aminophospholipid, phosphatidylserine (PS), is typically restricted to the inner leaflet of the plasma membrane under normal, healthy physiological conditions. PS is irreversibly externalized during apoptosis, where it serves as a signal for elimination by efferocytosis. PS is also reversibly and transiently externalized during cell activation such as platelet and immune cell activation. These events associated with physiological PS externalization are tightly controlled by the regulated activation of flippases and scramblases. Indeed, improper regulation of PS externalization results in thrombotic diseases such as Scott Syndrome, a defect in coagulation and thrombin production, and in the case of efferocytosis, can result in autoimmunity such as systemic lupus erythematosus (SLE) when PS-mediated apoptosis and efferocytosis fails. The physiological regulation of PS is also perturbed in cancer and during viral infection, whereby PS becomes persistently exposed on the surface of such stressed and diseased cells, which can lead to chronic thrombosis and chronic immune evasion. In this review, we summarize evidence for the dysregulation of PS with a main focus on cancer biology and the pathogenic mechanisms for immune evasion and signaling by PS, as well as the discussion of new therapeutic strategies aimed to target externalized PS. We posit that chronic PS externalization is a universal and agnostic marker for diseased tissues, and in cancer, likely reflects a cell intrinsic form of immune escape. The continued development of new therapeutic strategies for targeting PS also provides rationale for their co-utility as adjuvants and with immune checkpoint therapeutics.
Collapse
Affiliation(s)
- Rachael Pulica
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Ahmed Aquib
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Christopher Varsanyi
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Ziren Wang
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Trevor Frederick
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - David C Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Bhumik Patel
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Kenneth Vergel de Dios
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Victor Poalasin
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Mariana S De Lorenzo
- Department of Cell Biology and Molecular Medicine, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Sergei V Kotenko
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA
| | - Yi Wu
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Aizen Yang
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Alok Choudhary
- International Center for Public Health, Public Health Research Institute, Newark, NJ, 07103, USA
| | - Ganapathy Sriram
- Department Biological, Chemical and Environmental Sciences, Wheaton College, 26 E Main St, Norton, MA, 02766, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Center for Cell Signaling, Rutgers New Jersey Medical School, 205 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
7
|
Jan LY, Jan YN. Wide-ranging cellular functions of ion channels and lipid scramblases in the structurally related TMC, TMEM16 and TMEM63 families. Nat Struct Mol Biol 2025; 32:222-236. [PMID: 39715905 DOI: 10.1038/s41594-024-01444-x] [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: 01/18/2024] [Accepted: 10/31/2024] [Indexed: 12/25/2024]
Abstract
Calcium (Ca2+)-activated ion channels and lipid scramblases in the transmembrane protein 16 (TMEM16) family are structurally related to mechanosensitive ion channels in the TMEM63 and transmembrane channel-like (TMC) families. Members of this structurally related superfamily share similarities in gating transitions and serve a wide range of physiological functions, which is evident from their disease associations. The TMEM16, TMEM63 and TMC families include members with important functions in the cell membrane and/or intracellular organelles such as the endoplasmic reticulum, membrane contact sites, endosomes and lysosomes. Moreover, some members of the TMEM16 family and the TMC family perform dual functions of ion channel and lipid scramblase, leading to intriguing physiological implications. In addition to their physiological functions such as mediating phosphatidylserine exposure and facilitation of extracellular vesicle generation and cell fusion, scramblases are involved in the entry and replication of enveloped viruses. Comparisons of structurally diverse scramblases may uncover features in the lipid-scrambling mechanisms that are likely shared by scramblases.
Collapse
Affiliation(s)
- Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Yuh Nung Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Cohen‐Adiv S, Amer‐Sarsour F, Berdichevsky Y, Boxer E, Goldstein O, Gana‐Weisz M, Tripathi U, Rike WA, Prag G, Gurevich T, Giladi N, Stern S, Orr‐Urtreger A, Friedmann‐Morvinski D, Ashkenazi A. TMEM16F regulates pathologic α-synuclein secretion and spread in cellular and mouse models of Parkinson's disease. Aging Cell 2025; 24:e14387. [PMID: 39487963 PMCID: PMC11822650 DOI: 10.1111/acel.14387] [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: 04/15/2024] [Revised: 09/14/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024] Open
Abstract
One of the main hallmarks of Parkinson's disease (PD) pathology is the spread of the aggregate-prone protein α-synuclein (α-syn), which can be detected in the plasma and cerebrospinal fluid of patients as well as in the extracellular environment of neuronal cells. The secreted α-syn can exhibit "prion-like" behavior and transmission to naïve cells can promote conformational changes and pathology. The precise role of plasma membrane proteins in the pathologic process of α-syn is yet to be fully resolved. The TMEM16 family of lipid scramblases and ion channels has been recently associated with cancer and infectious diseases but is less known for its role in aging-related diseases. To elucidate the role of TMEM16F in α-syn spread, we transduced neurons derived from TMEM16F knockout mice with a reporter system that enables the distinction between donor and recipient neurons of pathologic α-synA53T. We found that the spread of α-synA53T was reduced in neurons derived from TMEM16F-knockout mice. These findings were recapitulated in vivo in a mouse model of PD, where attenuated α-synA53T spread was observed when TMEM16F was ablated. Moreover, we identified a single nucleotide polymorphism in TMEM16F of Ashkenazi Jewish PD patients resulting in a missense Ala703Ser mutation with enhanced lipid scramblase activity. This mutation is associated with altered regulation of α-synA53T extracellular secretion in cellular models of PD. Our study highlights TMEM16F as a novel regulator of α-syn spread and as a potential therapeutic target in synucleinopathies.
Collapse
Affiliation(s)
- Stav Cohen‐Adiv
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Fatima Amer‐Sarsour
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Yevgeny Berdichevsky
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Emily Boxer
- The School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Orly Goldstein
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Mali Gana‐Weisz
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Wote Amelo Rike
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Gali Prag
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Tanya Gurevich
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Division, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Nir Giladi
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Brain DivisionTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Avi Orr‐Urtreger
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Dinorah Friedmann‐Morvinski
- The School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Avraham Ashkenazi
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
9
|
Kim JE, Ko W, Jin S, Woo JN, Jung Y, Bae I, Choe HK, Seo D, Hille B, Suh BC. Activation of TMEM16E scramblase induces ligand independent growth factor receptor signaling and macropinocytosis for membrane repair. Commun Biol 2025; 8:35. [PMID: 39794444 PMCID: PMC11724107 DOI: 10.1038/s42003-025-07465-6] [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: 03/28/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The calcium-dependent phospholipid scramblase TMEM16E mediates ion transport and lipid translocation across the plasma membrane. TMEM16E also contributes to protection of membrane structure by facilitating cellular repair signaling. Our research reveals that TMEM16E activation promotes macropinocytosis, essential for maintaining plasma membrane integrity. This scramblase externalizes phosphatidylserine, typically linked to resting growth factor receptors. We demonstrate that TMEM16E can interact with and signal through growth factor receptors, including epidermal growth factor receptor, even without ligands. This interaction stimulates downstream phosphoinositide 3-kinase and facilitates macropinocytosis and internalization of annexin V bound to the membrane, a process sensitive to amiloride inhibition. Although TMEM16E is internalized during this process, it returns to the plasma membrane. TMEM16E- driven macropinocytosis is proposed to restore membrane integrity after perturbation, potentially explaining pathologies in conditions like muscular dystrophies, where TMEM16E functionality is compromised, highlighting its critical role in muscle cell survival.
Collapse
Affiliation(s)
- Jung-Eun Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Woori Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Siwoo Jin
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Jin-Nyeong Woo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Yuna Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Inah Bae
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Han-Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Bertil Hille
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
10
|
Dorca-Arévalo J, Santana-Ruiz A, Torrejón-Escribano B, Martín-Satué M, Blasi J. Epsilon Toxin from Clostridium perfringens Induces the Generation of Extracellular Vesicles in HeLa Cells Overexpressing Myelin and Lymphocyte Protein. Toxins (Basel) 2024; 16:525. [PMID: 39728783 PMCID: PMC11728497 DOI: 10.3390/toxins16120525] [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: 11/12/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Epsilon toxin (ETX) from Clostridium perfringens is a pore-forming toxin (PFT) that crosses the blood-brain barrier and binds to myelin structures. In in vitro assays, ETX causes oligodendrocyte impairment, subsequently leading to demyelination. In fact, ETX has been associated with triggering multiple sclerosis. Myelin and lymphocyte protein (MAL) is widely considered to be the receptor for ETX as its presence is crucial for the effects of ETX on the plasma membrane of host cells that involve pore formation, resulting in cell death. To overcome the pores formed by PFTs, some host cells produce extracellular vesicles (EVs) to reduce the amount of pores inserted into the plasma membrane. The formation of EVs has not been studied for ETX in host cells. Here, we generated a highly sensitive clone from HeLa cells overexpressing the MAL-GFP protein in the plasma membrane. We observed that ETX induces the formation of EVs. Moreover, the MAL protein and ETX oligomers are found in these EVs, which are a very useful tool to decipher and study the mode of action of ETX and characterize the mechanisms involved in the binding of ETX to its receptor.
Collapse
Affiliation(s)
- Jonatan Dorca-Arévalo
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, 08907 Barcelona, Spain; (A.S.-R.); (B.T.-E.); (M.M.-S.); (J.B.)
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Spain
- Institute of Neuroscience, Bellvitge Health Sciences Campus, University of Barcelona, Carrer de la Feixa Llarga, s/n, 08907 L’Hospitalet de Llobregat, Spain
| | - Antonio Santana-Ruiz
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, 08907 Barcelona, Spain; (A.S.-R.); (B.T.-E.); (M.M.-S.); (J.B.)
| | - Benjamín Torrejón-Escribano
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, 08907 Barcelona, Spain; (A.S.-R.); (B.T.-E.); (M.M.-S.); (J.B.)
- Scientific and Technological Centers (CCiTUB), Bellvitge Campus, University of Barcelona, Carrer de la Feixa Llarga, s/n, 08907 L’Hospitalet de Llobregat, Spain
| | - Mireia Martín-Satué
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, 08907 Barcelona, Spain; (A.S.-R.); (B.T.-E.); (M.M.-S.); (J.B.)
- Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), CIBERONC, 08908 L’Hospitalet de Llobregat, Spain
| | - Juan Blasi
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, 08907 Barcelona, Spain; (A.S.-R.); (B.T.-E.); (M.M.-S.); (J.B.)
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Spain
- Institute of Neuroscience, Bellvitge Health Sciences Campus, University of Barcelona, Carrer de la Feixa Llarga, s/n, 08907 L’Hospitalet de Llobregat, Spain
| |
Collapse
|
11
|
Huang Z, Iqbal Z, Zhao Z, Chen X, Mahmmod A, Liu J, Li W, Deng Z. TMEM16 proteins: Ca 2+‑activated chloride channels and phospholipid scramblases as potential drug targets (Review). Int J Mol Med 2024; 54:81. [PMID: 39092585 PMCID: PMC11315658 DOI: 10.3892/ijmm.2024.5405] [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: 03/26/2024] [Accepted: 06/06/2024] [Indexed: 08/04/2024] Open
Abstract
TMEM16 proteins, which function as Ca2+‑activated Cl‑ channels are involved in regulating a wide variety of cellular pathways and functions. The modulators of Cl‑ channels can be used for the molecule‑based treatment of respiratory diseases, cystic fibrosis, tumors, cancer, osteoporosis and coronavirus disease 2019. The TMEM16 proteins link Ca2+ signaling, cellular electrical activity and lipid transport. Thus, deciphering these complex regulatory mechanisms may enable a more comprehensive understanding of the physiological functions of the TMEM16 proteins and assist in ascertaining the applicability of these proteins as potential pharmacological targets for the treatment of a range of diseases. The present review examined the structures, functions and characteristics of the different types of TMEM16 proteins, their association with the pathogenesis of various diseases and the applicability of TMEM16 modulator‑based treatment methods.
Collapse
Affiliation(s)
- Zeqi Huang
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zoya Iqbal
- Department of Orthopaedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhe Zhao
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Xiaoqiang Chen
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Ayesha Mahmmod
- Faculty of Pharmacy, The University of Lahore, Lahore, Punjab 58240, Pakistan
| | - Jianquan Liu
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Wencui Li
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| | - Zhiqin Deng
- Department of Hand and Foot Surgery, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
12
|
Cases M, Dorca-Arévalo J, Blanch M, Rodil S, Terni B, Martín-Satué M, Llobet A, Blasi J, Solsona C. The epsilon toxin from Clostridium perfringens stimulates calcium-activated chloride channels, generating extracellular vesicles in Xenopus oocytes. Pharmacol Res Perspect 2024; 12:e70005. [PMID: 39320019 PMCID: PMC11423345 DOI: 10.1002/prp2.70005] [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] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 09/26/2024] Open
Abstract
The epsilon toxin (Etx) from Clostridium perfringens has been identified as a potential trigger of multiple sclerosis, functioning as a pore-forming toxin that selectively targets cells expressing the plasma membrane (PM) myelin and lymphocyte protein (MAL). Previously, we observed that Etx induces the release of intracellular ATP in sensitive cell lines. Here, we aimed to re-examine the mechanism of action of the toxin and investigate the connection between pore formation and ATP release. We examined the impact of Etx on Xenopus laevis oocytes expressing human MAL. Extracellular ATP was assessed using the luciferin-luciferase reaction. Activation of calcium-activated chloride channels (CaCCs) and a decrease in the PM surface were recorded using the two-electrode voltage-clamp technique. To evaluate intracellular Ca2+ levels and scramblase activity, fluorescent dyes were employed. Extracellular vesicles were imaged using light and electron microscopy, while toxin oligomers were identified through western blots. Etx triggered intracellular Ca2+ mobilization in the Xenopus oocytes expressing hMAL, leading to the activation of CaCCs, ATP release, and a reduction in PM capacitance. The toxin induced the activation of scramblase and, thus, translocated phospholipids from the inner to the outer leaflet of the PM, exposing phosphatidylserine outside in Xenopus oocytes and in an Etx-sensitive cell line. Moreover, Etx caused the formation of extracellular vesicles, not derived from apoptotic bodies, through PM fission. These vesicles carried toxin heptamers and doughnut-like structures in the nanometer size range. In conclusion, ATP release was not directly attributed to the formation of pores in the PM, but to scramblase activity and the formation of extracellular vesicles.
Collapse
Affiliation(s)
- Mercè Cases
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Jonatan Dorca-Arévalo
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Marta Blanch
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Sergi Rodil
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
| | - Beatrice Terni
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Mireia Martín-Satué
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
| | - Artur Llobet
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Juan Blasi
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Carles Solsona
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Tang J, Song H, Li S, Lam SM, Ping J, Yang M, Li N, Chang T, Yu Z, Liu W, Lu Y, Zhu M, Tang Z, Liu Z, Guo YR, Shui G, Veillette A, Zeng Z, Wu N. TMEM16F Expressed in Kupffer Cells Regulates Liver Inflammation and Metabolism to Protect Against Listeria Monocytogenes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402693. [PMID: 39136057 PMCID: PMC11497084 DOI: 10.1002/advs.202402693] [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: 03/14/2024] [Revised: 06/20/2024] [Indexed: 10/25/2024]
Abstract
Infection by bacteria leads to tissue damage and inflammation, which need to be tightly controlled by host mechanisms to avoid deleterious consequences. It is previously reported that TMEM16F, a calcium-activated lipid scramblase expressed in various immune cell types including T cells and neutrophils, is critical for the control of infection by bacterium Listeria monocytogenes (Lm) in vivo. This function correlated with the capacity of TMEM16F to repair the plasma membrane (PM) damage induced in T cells in vitro, by the Lm toxin listeriolysin O (LLO). However, whether the protective effect of TMEM16F on Lm infection in vivo is mediated by an impact in T cells, or in other cell types, is not determined. Herein, the immune cell types and mechanisms implicated in the protective effect of TMEM16F against Lm in vivo are elucidated. Cellular protective effects of TMEM16F correlated with its capacity of lipid scrambling and augment PM fluidity. Using cell type-specific TMEM16F-deficient mice, the indication is obtained that TMEM16F expressed in liver Kupffer cells (KCs), but not in T cells or B cells, is key for protection against Listeria in vivo. In the absence of TMEM16F, Listeria induced PM rupture and fragmentation of KCs in vivo. KC death associated with greater liver damage, inflammatory changes, and dysregulated liver metabolism. Overall, the results uncovered that TMEM16F expressed in Kupffer cells is crucial to protect the host against Listeria infection. This influence is associated with the capacity of Kupffer cell-expressed TMEM16F to prevent excessive inflammation and abnormal liver metabolism.
Collapse
Affiliation(s)
- Jianlong Tang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Hua Song
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
| | - Shimin Li
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - Jieming Ping
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Mengyun Yang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Na Li
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Teding Chang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ze Yu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Weixiang Liu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Yan Lu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Min Zhu
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhaohui Tang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Yusong R. Guo
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - André Veillette
- Laboratory of Molecular OncologyInstitut de recherches cliniques de Montréal (IRCM)MontréalQuébecH2W1R7Canada
- Department of MedicineUniversity of MontréalMontréalQuébecH3T 1J4Canada
- Department of MedicineMcGill UniversityMontréalQuébecH3G 1Y6Canada
| | - Zhutian Zeng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Department of OncologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefei230001China
| | - Ning Wu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
14
|
Williams JK, Ngo JM, Murugupandiyan A, Croall DE, Hartzell HC, Schekman R. Calpains Orchestrate Secretion of Annexin-containing Microvesicles during Membrane Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611512. [PMID: 39282443 PMCID: PMC11398502 DOI: 10.1101/2024.09.05.611512] [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: 09/21/2024]
Abstract
Microvesicles (MVs) are membrane-enclosed, plasma membrane-derived particles released by cells from all branches of life. MVs have utility as disease biomarkers and may participate in intercellular communication; however, physiological processes that induce their secretion are not known. Here, we isolate and characterize annexin-containing MVs and show that these vesicles are secreted in response to the calcium influx caused by membrane damage. The annexins in these vesicles are cleaved by calpains. After plasma membrane injury, cytoplasmic calcium-bound annexins are rapidly recruited to the plasma membrane and form a scab-like structure at the lesion. In a second phase, recruited annexins are cleaved by calpains-1/2, disabling membrane scabbing. Cleavage promotes annexin secretion within MVs. Our data supports a new model of plasma membrane repair, where calpains relax annexin-membrane aggregates in the lesion repair scab, allowing secretion of damaged membrane and annexins as MVs. We anticipate that cells experiencing plasma membrane damage, including muscle and metastatic cancer cells, secrete these MVs at elevated levels.
Collapse
Affiliation(s)
- Justin Krish Williams
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Jordan Matthew Ngo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Abinayaa Murugupandiyan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Dorothy E. Croall
- Department of Biochemistry, Microbiology and Molecular Biology, University of Maine, Orono, United States
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - Randy Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
15
|
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
|
16
|
Shan KZ, Le T, Liang P, Dong P, Lowry AJ, Kremmyda P, Claesson-Welsh L, Yang H. TMEM16F scramblase regulates angiogenesis via endothelial intracellular signaling. J Cell Sci 2024; 137:jcs261566. [PMID: 38940198 PMCID: PMC11273297 DOI: 10.1242/jcs.261566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
TMEM16F (also known as ANO6), a Ca2+-activated lipid scramblase (CaPLSase) that dynamically disrupts lipid asymmetry, plays a crucial role in various physiological and pathological processes, such as blood coagulation, neurodegeneration, cell-cell fusion and viral infection. However, the mechanisms through which it regulates these processes remain largely elusive. Using endothelial cell-mediated angiogenesis as a model, here we report a previously unknown intracellular signaling function of TMEM16F. We demonstrate that TMEM16F deficiency impairs developmental retinal angiogenesis in mice and disrupts angiogenic processes in vitro. Biochemical analyses indicate that the absence of TMEM16F enhances the plasma membrane association of activated Src kinase. This in turn increases VE-cadherin phosphorylation and downregulation, accompanied by suppressed angiogenesis. Our findings not only highlight the role of intracellular signaling by TMEM16F in endothelial cells but also open new avenues for exploring the regulatory mechanisms for membrane lipid asymmetry and their implications in disease pathogenesis.
Collapse
Affiliation(s)
- Ke Zoe Shan
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Trieu Le
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Ping Dong
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Polina Kremmyda
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck, Beijer and SciLifeLab Laboratory, Uppsala 751 85, Sweden
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck, Beijer and SciLifeLab Laboratory, Uppsala 751 85, Sweden
| | - Huanghe Yang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
- Department of Neurobiology, Duke University, School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
17
|
Genovese M, Galietta LJV. Anoctamin pharmacology. Cell Calcium 2024; 121:102905. [PMID: 38788257 DOI: 10.1016/j.ceca.2024.102905] [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: 03/21/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024]
Abstract
TMEM16 proteins, also known as anoctamins, are a family of ten membrane proteins with various tissue expression and subcellular localization. TMEM16A (anoctamin 1) is a plasma membrane protein that acts as a calcium-activated chloride channel. It is expressed in many types of epithelial cells, smooth muscle cells and some neurons. In airway epithelial cells, TMEM16A expression is particularly enhanced by inflammatory stimuli that also promote goblet cell metaplasia and mucus hypersecretion. Therefore, pharmacological modulation of TMEM16A could be beneficial to improve mucociliary clearance in chronic obstructive respiratory diseases. However, the correct approach to modulate TMEM16A activity (activation or inhibition) is still debated. Pharmacological inhibitors of TMEM16A could also be useful as anti-hypertensive agents given the TMEM16A role in smooth muscle contraction. In contrast to TMEM16A, TMEM16F (anoctamin 6) behaves as a calcium-activated phospholipid scramblase, responsible for the externalization of phosphatidylserine on cell surface. Inhibitors of TMEM16F could be useful as anti-coagulants and anti-viral agents. The role of other anoctamins as therapeutic targets is still unclear since their physiological role is still to be defined.
Collapse
Affiliation(s)
- Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy; Department of Translational Medical Sciences (DISMET), University of Naples "Federico II", Italy.
| |
Collapse
|
18
|
Yan M, Wang Z, An Y, Li Z, Li Y, Zhang H, Li C, Wang L, Chen L, Gao C, Wang D, Gao C. OxLDL enhances procoagulant activity of endothelial cells by TMEM16F-mediated phosphatidylserine exposure. Cell Biol Int 2024; 48:848-860. [PMID: 38444077 DOI: 10.1002/cbin.12150] [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: 07/10/2023] [Revised: 01/28/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
Oxidized low-density lipoprotein (oxLDL), a key component in atherosclerosis and hyperlipidemia, is a risk factor for atherothrombosis in dyslipidemia, yet its mechanism is poorly understood. In this study, we used oxLDL-induced human aortic endothelial cells (HAECs) and high-fat diet (HFD)-fed mice as a hyperlipidemia model. Phosphatidylserine (PS) exposure, cytosolic Ca2+, reactive oxygen species (ROS), and lipid peroxidation were measured by flow cytometer. TMEM16F expression was detected by immunofluorescence, western blot, and reverse transcription polymerase chain reaction. Procoagulant activity (PCA) was measured by coagulation time, intrinsic/extrinsic factor Xase, and thrombin generation. We found that oxLDL-induced PS exposure and the corresponding PCA of HAECs were increased significantly compared with control, which could be inhibited over 90% by lactadherin. Importantly, TMEM16F expression in oxLDL-induced HAECs was upregulated by enhanced intracellular Ca2+ concentration, ROS, and lipid peroxidation, which led to PS exposure. Meanwhile, the knockdown of TMEM16F by short hairpin RNA significantly inhibited PS exposure in oxLDL-induced HAECs. Moreover, we observed that HFD-fed mice dramatically increased the progress of thrombus formation and accompanied upregulated TMEM16F expression by thromboelastography analysis, FeCl3-induced carotid artery thrombosis model, and western blot. Collectively, these results demonstrate that TMEM16F-mediated PS exposure may contribute to prothrombotic status under hyperlipidemic conditions, which may serve as a novel therapeutic target for the prevention of thrombosis in hyperlipidemia.
Collapse
Affiliation(s)
- Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Zelong Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Zhanni Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Yun Li
- Hematology Department, Daqing Oil Field General Hospital, Daqing, China
| | - Hongyu Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Caixia Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Lifeng Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Li Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Chao Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Dongsheng Wang
- Department of Emergency, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| |
Collapse
|
19
|
He Z, Liu D, Li H, Gao W, Li X, Ma H, Shi W. Amphiphilic Rhodamine Fluorescent Probes Combined with Basal Imaging for Fine Structures of the Cell Membrane. Anal Chem 2024; 96:7257-7264. [PMID: 38664861 DOI: 10.1021/acs.analchem.4c00946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Confocal fluorescence imaging of fine structures of the cell membrane is important for understanding their biofunctions but is often neglected due to the lack of an effective method. Herein, we develop new amphiphilic rhodamine fluorescent probe RMGs in combination with basal imaging for this purpose. The probes show high signal-to-noise ratio and brightness and low internalization rate, making them suitable for imaging the fine substructures of the cell membrane. Using the representative probe RMG3, we not only observed the cell pseudopodia and intercellular nanotubes but also monitored the formation of migrasomes in real time. More importantly, in-depth imaging studies on more cell lines revealed for the first time that hepatocellular carcinoma cells secreted much more adherent extracellular vesicles than other cell lines, which might serve as a potential indicator of liver cells. We believe that RMGs may be useful for investigating the fine structures of the cell membrane.
Collapse
Affiliation(s)
- Zixu He
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Diankai Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - He Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenjie Gao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohua Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Huimin Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen Shi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Suda K, Moriyama Y, Razali N, Chiu Y, Masukagami Y, Nishimura K, Barbee H, Takase H, Sugiyama S, Yamazaki Y, Sato Y, Higashiyama T, Johmura Y, Nakanishi M, Kono K. Plasma membrane damage limits replicative lifespan in yeast and induces premature senescence in human fibroblasts. NATURE AGING 2024; 4:319-335. [PMID: 38388781 PMCID: PMC10950784 DOI: 10.1038/s43587-024-00575-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
Plasma membrane damage (PMD) occurs in all cell types due to environmental perturbation and cell-autonomous activities. However, cellular outcomes of PMD remain largely unknown except for recovery or death. In this study, using budding yeast and normal human fibroblasts, we found that cellular senescence-stable cell cycle arrest contributing to organismal aging-is the long-term outcome of PMD. Our genetic screening using budding yeast unexpectedly identified a close genetic association between PMD response and replicative lifespan regulations. Furthermore, PMD limits replicative lifespan in budding yeast; upregulation of membrane repair factors ESCRT-III (SNF7) and AAA-ATPase (VPS4) extends it. In normal human fibroblasts, PMD induces premature senescence via the Ca2+-p53 axis but not the major senescence pathway, DNA damage response pathway. Transient upregulation of ESCRT-III (CHMP4B) suppressed PMD-dependent senescence. Together with mRNA sequencing results, our study highlights an underappreciated but ubiquitous senescent cell subtype: PMD-dependent senescent cells.
Collapse
Affiliation(s)
- Kojiro Suda
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yohsuke Moriyama
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Nurhanani Razali
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yatzu Chiu
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yumiko Masukagami
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Hyogo, Japan
| | - Hunter Barbee
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Hiroshi Takase
- Core Laboratory, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Shinju Sugiyama
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yuta Yamazaki
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yoshikatsu Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Tetsuya Higashiyama
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Yoshikazu Johmura
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Keiko Kono
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
21
|
Kim E, Bang J, Sung JH, Lee J, Shin DH, Kim S, Lee BC. Generation of human TMEM16F-specific affibodies using purified TMEM16F. Front Mol Biosci 2024; 10:1319251. [PMID: 38274091 PMCID: PMC10808743 DOI: 10.3389/fmolb.2023.1319251] [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: 10/19/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: TMEM16 family proteins are involved in a variety of functions, including ion transport, phospholipid scrambling, and the regulation of membrane proteins. Among them, TMEM16F has dual functions as a phospholipid scramblase and a nonselective ion channel. TMEM16F is widely expressed and functions in platelet activation during blood clotting, bone formation, and T cell activation. Despite the functional importance of TMEM16F, the modulators of TMEM16F function have not been sufficiently studied. Method: In this study, we generated TMEM16F-specific affibodies by performing phage display with brain-specific TMEM16F (hTMEM16F) variant 1 purified from GnTi- cells expressing this variant in the presence of digitonin as a detergent. Purified human TMEM16F protein, which was proficient in transporting phospholipids in a Ca2+-dependent manner in proteoliposomes, was coated onto plates and then the phage library was added to fish out TMEM16F-binding affibodies. For the validation of interaction between affibodies and TMEM16F proteins, ELISA, bio-layer interferometry, and size exclusion chromatography were conducted. Results and Discussion: As a result, the full sequences of 38 candidates were acquired from 98 binding candidates. Then, we selected 10 candidates and purified seven of them from E. coli expressing these candidates. Using various assays, we confirmed that two affibodies bound to human TMEM16F with high affinity. These affibodies can be useful for therapeutical and diagnostic applications of TMEM16F-related cancer and neurodegenerative diseases. Future studies will be required to investigate the effects of these affibodies on TMEM16F function.
Collapse
Affiliation(s)
- Eunyoung Kim
- Korea Brain Research Institute, Neurovascular Unit Research Group, Daegu, Republic of Korea
| | - Jinho Bang
- Korea Institute of Ceramic Engineering and Technology, Bio-Healthcare Materials Center, Cheongju, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Ji Hye Sung
- Korea Brain Research Institute, Neurovascular Unit Research Group, Daegu, Republic of Korea
| | - Jonghwan Lee
- Korea Institute of Ceramic Engineering and Technology, Bio-Healthcare Materials Center, Cheongju, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Sunghyun Kim
- Korea Institute of Ceramic Engineering and Technology, Bio-Healthcare Materials Center, Cheongju, Republic of Korea
| | - Byoung-Cheol Lee
- Korea Brain Research Institute, Neurovascular Unit Research Group, Daegu, Republic of Korea
| |
Collapse
|
22
|
Ye Z, Galvanetto N, Puppulin L, Pifferi S, Flechsig H, Arndt M, Triviño CAS, Di Palma M, Guo S, Vogel H, Menini A, Franz CM, Torre V, Marchesi A. Structural heterogeneity of the ion and lipid channel TMEM16F. Nat Commun 2024; 15:110. [PMID: 38167485 PMCID: PMC10761740 DOI: 10.1038/s41467-023-44377-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Transmembrane protein 16 F (TMEM16F) is a Ca2+-activated homodimer which functions as an ion channel and a phospholipid scramblase. Despite the availability of several TMEM16F cryogenic electron microscopy (cryo-EM) structures, the mechanism of activation and substrate translocation remains controversial, possibly due to restrictions in the accessible protein conformational space. In this study, we use atomic force microscopy under physiological conditions to reveal a range of structurally and mechanically diverse TMEM16F assemblies, characterized by variable inter-subunit dimerization interfaces and protomer orientations, which have escaped prior cryo-EM studies. Furthermore, we find that Ca2+-induced activation is associated to stepwise changes in the pore region that affect the mechanical properties of transmembrane helices TM3, TM4 and TM6. Our direct observation of membrane remodelling in response to Ca2+ binding along with additional electrophysiological analysis, relate this structural multiplicity of TMEM16F to lipid and ion permeation processes. These results thus demonstrate how conformational heterogeneity of TMEM16F directly contributes to its diverse physiological functions.
Collapse
Affiliation(s)
- Zhongjie Ye
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Nicola Galvanetto
- Department of Physics, University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland
| | - Leonardo Puppulin
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, I-30172 Mestre, Venice, Italy
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Simone Pifferi
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Holger Flechsig
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Melanie Arndt
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland
| | | | - Michael Di Palma
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Shifeng Guo
- Shenzhen Key Laboratory of Smart Sensing and Intelligent Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Horst Vogel
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- Institut des Sciences et Ingénierie Chimiques (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Anna Menini
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan
| | - Vincent Torre
- International School for Advanced Studies (SISSA), 34136, Trieste, Italy.
- Institute of Materials (ION-CNR), Area Science Park, Basovizza, 34149, Trieste, Italy.
- BIoValley Investments System and Solutions (BISS), 34148, Trieste, Italy.
| | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-machi, 920-1192, Kanazawa, Japan.
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.
| |
Collapse
|
23
|
Fang J, Zhang Y, Zhu T, Li Y. Scramblase activity of proteorhodopsin confers physiological advantages to Escherichia coli in the absence of light. iScience 2023; 26:108551. [PMID: 38125024 PMCID: PMC10730872 DOI: 10.1016/j.isci.2023.108551] [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: 02/03/2023] [Revised: 05/11/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Microbial rhodopsins are widely distributed in the aqua-ecosystem due to their simple structure and multifaceted functions. Conventionally, microbial rhodopsins are considered to be exclusively light active. Here, we report the discovery of light-independent function of a proteorhodopsin from a psychrophile Psychroflexus torquis (ptqPR). ptqPR could improve the growth and viability of Escherichia coli cells under stressful conditions in the absence of light, and this was achieved by improving the energy maintenance, membrane potential, membrane fluidity, and membrane integrity. We further show that this non-canonical function of PR is related to its scramblase activity. PR mutants which lost scramblase activities also lost their ability to confer physiological advantages in E. coli. These findings shed light on why microbial rhodopsins are widely distributed in ecological systems where light is inaccessible.
Collapse
Affiliation(s)
- Jiayu Fang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanping Zhang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Taicheng Zhu
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yin Li
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
24
|
Wu N, Veillette A. Lipid scrambling in immunology: why it is important. Cell Mol Immunol 2023; 20:1081-1083. [PMID: 37012395 PMCID: PMC10071233 DOI: 10.1038/s41423-023-01009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023] Open
Affiliation(s)
- Ning Wu
- The First Affiliated Hospital of Anhui Medical University, Clinical Immunology Institute, Anhui Medical University, Hefei, Anhui, China.
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, H2W1R7, Canada.
- Department of Medicine, University of Montréal, Montréal, QC, H3T 1J4, Canada.
- Department of Medicine, McGill University, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
25
|
Shan KZ, Le T, Liang P, Dong P, Yang H. Endothelial TMEM16F lipid scramblase regulates angiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553724. [PMID: 37645870 PMCID: PMC10462142 DOI: 10.1101/2023.08.17.553724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Dynamic loss of lipid asymmetry through the activation of TMEM16 Ca2+-activated lipid scramblases (CaPLSases) has been increasingly recognized as an essential membrane event in a wide range of physiological and pathological processes, including blood coagulation, microparticle release, bone development, pain sensation, cell-cell fusion, and viral infection. Despite the recent implications of TMEM16F CaPLSase in vascular development and endothelial cell-mediated coagulation, its signaling role in endothelial biology remains to be established. Here, we show that endothelial TMEM16F regulates in vitro and in vivo angiogenesis through intracellular signaling. Developmental retinal angiogenesis is significantly impaired in TMEM16F deficient mice, as evidenced by fewer vascular loops and larger loop areas. Consistent with our in vivo observation, TMEM16F siRNA knockdown in human umbilical vein endothelial cells compromises angiogenesis in vitro. We further discovered that TMEM16F knockdown enhances VE-cadherin phosphorylation and reduces its expression. Moreover, TMEM16F knockdown also promotes Src kinase phosphorylation at tyrosine 416, which may be responsible for downregulating VE-cadherin expression. Our study thus uncovers a new biological function of TMEM16F in angiogenesis and provides a potential mechanism for how the CaPLSase regulates angiogenesis through intracellular signaling.
Collapse
Affiliation(s)
- Ke Zoe Shan
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Trieu Le
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
- Curreent address: Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Pengfei Liang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Ping Dong
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University, School of Medicine, Durham, NC 27710, USA
- Curreent address: Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| |
Collapse
|
26
|
Feng S, Puchades C, Ko J, Wu H, Chen Y, Figueroa EE, Gu S, Han TW, Ho B, Cheng T, Li J, Shoichet B, Jan YN, Cheng Y, Jan LY. Identification of a drug binding pocket in TMEM16F calcium-activated ion channel and lipid scramblase. Nat Commun 2023; 14:4874. [PMID: 37573365 PMCID: PMC10423226 DOI: 10.1038/s41467-023-40410-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 07/23/2023] [Indexed: 08/14/2023] Open
Abstract
The dual functions of TMEM16F as Ca2+-activated ion channel and lipid scramblase raise intriguing questions regarding their molecular basis. Intrigued by the ability of the FDA-approved drug niclosamide to inhibit TMEM16F-dependent syncytia formation induced by SARS-CoV-2, we examined cryo-EM structures of TMEM16F with or without bound niclosamide or 1PBC, a known blocker of TMEM16A Ca2+-activated Cl- channel. Here, we report evidence for a lipid scrambling pathway along a groove harboring a lipid trail outside the ion permeation pore. This groove contains the binding pocket for niclosamide and 1PBC. Mutations of two residues in this groove specifically affect lipid scrambling. Whereas mutations of some residues in the binding pocket of niclosamide and 1PBC reduce their inhibition of TMEM16F-mediated Ca2+ influx and PS exposure, other mutations preferentially affect the ability of niclosamide and/or 1PBC to inhibit TMEM16F-mediated PS exposure, providing further support for separate pathways for ion permeation and lipid scrambling.
Collapse
Affiliation(s)
- Shengjie Feng
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Cristina Puchades
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Juyeon Ko
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Hao Wu
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Yifei Chen
- Howard Hughes Medical Institute; UCSF, San Francisco, CA, USA
| | - Eric E Figueroa
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Shuo Gu
- BioDuro-Sundia Inc., Irvine, CA, USA
| | - Tina W Han
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- Dewpoint Therapeutics, Boston, MA, USA
| | - Brandon Ho
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Tong Cheng
- Howard Hughes Medical Institute; UCSF, San Francisco, CA, USA
| | - Junrui Li
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Brian Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF) School of Pharmacy, San Francisco, CA, USA
| | - Yuh Nung Jan
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- Howard Hughes Medical Institute; UCSF, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA.
- Howard Hughes Medical Institute; UCSF, San Francisco, CA, USA.
| | - Lily Yeh Jan
- Department of Physiology, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA.
- Howard Hughes Medical Institute; UCSF, San Francisco, CA, USA.
| |
Collapse
|
27
|
Anari M, Montgomery MK. Phospholipid metabolism in the liver - Implications for phosphatidylserine in non-alcoholic fatty liver disease. Biochem Pharmacol 2023; 213:115621. [PMID: 37217141 DOI: 10.1016/j.bcp.2023.115621] [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: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Mammalian cells contain more than a thousand different glycerophospholipid species that are essential membrane components and signalling molecules, with phosphatidylserine (PS) giving membranes their negative surface charge. Depending on the tissue, PS is important in apoptosis, blood clotting, cancer pathogenesis, as well as muscle and brain function, processes that are dependent on the asymmetrical distribution of PS on the plasma membrane and/or the capacity of PS to act as anchorage for various signalling proteins. Recent studies have implicated hepatic PS in the progression of non-alcoholic fatty liver disease (NAFLD), either as beneficial in the context of suppressing hepatic steatosis and fibrosis, or on the other hand as a potential contributor to the progression of liver cancer. This review provides an extensive overview of hepatic phospholipid metabolism, including its biosynthetic pathways, intracellular trafficking and roles in health and disease, further taking a deeper dive into PS metabolism, including associate and causative evidence of the role of PS in advanced liver disease.
Collapse
Affiliation(s)
- Marziyeh Anari
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
28
|
Dutta D. Interplay between membrane proteins and membrane protein-lipid pertaining to plant salinity stress. Cell Biochem Funct 2023. [PMID: 37158622 DOI: 10.1002/cbf.3798] [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: 11/26/2022] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
High salinity in agricultural lands is one of the predominant issues limiting agricultural yields. Plants have developed several mechanisms to withstand salinity stress, but the mechanisms are not effective enough for most crops to prevent and persist the salinity stress. Plant salt tolerance pathways involve membrane proteins that have a crucial role in sensing and mitigating salinity stress. Due to a strategic location interfacing two distinct cellular environments, membrane proteins can be considered checkpoints to the salt tolerance pathways in plants. Related membrane proteins functions include ion homeostasis, osmosensing or ion sensing, signal transduction, redox homeostasis, and small molecule transport. Therefore, modulating plant membrane proteins' function, expression, and distribution can improve plant salt tolerance. This review discusses the membrane protein-protein and protein-lipid interactions related to plant salinity stress. It will also highlight the finding of membrane protein-lipid interactions from the context of recent structural evidence. Finally, the importance of membrane protein-protein and protein-lipid interaction is discussed, and a future perspective on studying the membrane protein-protein and protein-lipid interactions to develop strategies for improving salinity tolerance is proposed.
Collapse
Affiliation(s)
- Debajyoti Dutta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| |
Collapse
|
29
|
Sakuragi T, Nagata S. Regulation of phospholipid distribution in the lipid bilayer by flippases and scramblases. Nat Rev Mol Cell Biol 2023:10.1038/s41580-023-00604-z. [PMID: 37106071 PMCID: PMC10134735 DOI: 10.1038/s41580-023-00604-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/29/2023]
Abstract
Cellular membranes function as permeability barriers that separate cells from the external environment or partition cells into distinct compartments. These membranes are lipid bilayers composed of glycerophospholipids, sphingolipids and cholesterol, in which proteins are embedded. Glycerophospholipids and sphingolipids freely move laterally, whereas transverse movement between lipid bilayers is limited. Phospholipids are asymmetrically distributed between membrane leaflets but change their location in biological processes, serving as signalling molecules or enzyme activators. Designated proteins - flippases and scramblases - mediate this lipid movement between the bilayers. Flippases mediate the confined localization of specific phospholipids (phosphatidylserine (PtdSer) and phosphatidylethanolamine) to the cytoplasmic leaflet. Scramblases randomly scramble phospholipids between leaflets and facilitate the exposure of PtdSer on the cell surface, which serves as an important signalling molecule and as an 'eat me' signal for phagocytes. Defects in flippases and scramblases cause various human diseases. We herein review the recent research on the structure of flippases and scramblases and their physiological roles. Although still poorly understood, we address the mechanisms by which they translocate phospholipids between lipid bilayers and how defects cause human diseases.
Collapse
Affiliation(s)
- Takaharu Sakuragi
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shigekazu Nagata
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
30
|
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
|
31
|
Barisch C, Holthuis JCM, Cosentino K. Membrane damage and repair: a thin line between life and death. Biol Chem 2023; 404:467-490. [PMID: 36810295 DOI: 10.1515/hsz-2022-0321] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/03/2023] [Indexed: 02/24/2023]
Abstract
Bilayered membranes separate cells from their surroundings and form boundaries between intracellular organelles and the cytosol. Gated transport of solutes across membranes enables cells to establish vital ion gradients and a sophisticated metabolic network. However, an advanced compartmentalization of biochemical reactions makes cells also particularly vulnerable to membrane damage inflicted by pathogens, chemicals, inflammatory responses or mechanical stress. To avoid potentially lethal consequences of membrane injuries, cells continuously monitor the structural integrity of their membranes and readily activate appropriate pathways to plug, patch, engulf or shed the damaged membrane area. Here, we review recent insights into the cellular mechanisms that underly an effective maintenance of membrane integrity. We discuss how cells respond to membrane lesions caused by bacterial toxins and endogenous pore-forming proteins, with a primary focus on the intimate crosstalk between membrane proteins and lipids during wound formation, detection and elimination. We also discuss how a delicate balance between membrane damage and repair determines cell fate upon bacterial infection or activation of pro-inflammatory cell death pathways.
Collapse
Affiliation(s)
- Caroline Barisch
- Molecular Infection Biology Division, Department of Biology and Center of Cellular Nanoanalytics, Osnabrück University, D-49076 Osnabrück, Germany
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology and Center of Cellular Nanoanalytics, Osnabrück University, D-49076 Osnabrück, Germany
| | - Katia Cosentino
- Molecular Cell Biophysics Division, Department of Biology and Center of Cellular Nanoanalytics, Osnabrück University, D-49076 Osnabrück, Germany
| |
Collapse
|
32
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
33
|
Dewdney B, Ursich L, Fletcher EV, Johns TG. Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma? Cancers (Basel) 2022; 14:cancers14235932. [PMID: 36497413 PMCID: PMC9740065 DOI: 10.3390/cancers14235932] [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: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is the most common form of high-grade glioma in adults and has a poor survival rate with very limited treatment options. There have been no significant advancements in glioblastoma treatment in over 30 years. Epidermal growth factor receptor is upregulated in most glioblastoma tumours and, therefore, has been a drug target in recent targeted therapy clinical trials. However, while many inhibitors and antibodies for epidermal growth factor receptor have demonstrated promising anti-tumour effects in preclinical models, they have failed to improve outcomes for glioblastoma patients in clinical trials. This is likely due to the highly plastic nature of glioblastoma tumours, which results in therapeutic resistance. Ion channels are instrumental in the development of many cancers and may regulate cellular plasticity in glioblastoma. This review will explore the potential involvement of a class of calcium-activated chloride channels called anoctamins in brain cancer. We will also discuss the integrated role of calcium channels and anoctamins in regulating calcium-mediated signalling pathways, such as epidermal growth factor signalling, to promote brain cancer cell growth and migration.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6319-1023
| | - Lauren Ursich
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| | - Terrance G. Johns
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
34
|
Taslimi A, Fields KM, Dahl KD, Liu Q, Tucker CL. Spatiotemporal control of necroptotic cell death and plasma membrane recruitment using engineered MLKL domains. Cell Death Dis 2022; 8:469. [PMID: 36446770 PMCID: PMC9709077 DOI: 10.1038/s41420-022-01258-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Necroptosis is a form of programmed necrotic cell death in which a signaling cascade induces oligomerization of mixed lineage kinase domain-like (MLKL) protein, leading to plasma membrane rupture. Necroptotic cell death is recognized as important for protection against viral infection and has roles in a variety of diseases, including cancer and diabetes. Despite its relevance to health and disease states, many questions remain about the precise mechanism of necroptotic cell death, cellular factors that can protect cells from necroptosis, and the role of necroptosis in disease models. In this study, we engineered a light-activated version of MLKL that rapidly oligomerizes and is recruited to the plasma membrane in cells exposed to light, inducing rapid cell death. We demonstrate this tool can be controlled spatially and temporally, used in a chemical genetic screen to identify chemicals and pathways that protect cells from MLKL-induced cell death, and used to study signaling responses of non-dying bystander cells. In additional studies, we re-engineered MLKL to block its cell-killing capacity but retain light-mediated membrane recruitment, developing a new single-component optogenetic tool that allows modulation of protein function at the plasma membrane.
Collapse
Affiliation(s)
- Amir Taslimi
- grid.430503.10000 0001 0703 675XDepartment of Pharmacology, Box 8303, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Kaiah M. Fields
- grid.430503.10000 0001 0703 675XDepartment of Pharmacology, Box 8303, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Kristin D. Dahl
- grid.430503.10000 0001 0703 675XDepartment of Pharmacology, Box 8303, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Qi Liu
- grid.430503.10000 0001 0703 675XDepartment of Pharmacology, Box 8303, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Chandra L. Tucker
- grid.430503.10000 0001 0703 675XDepartment of Pharmacology, Box 8303, University of Colorado School of Medicine, Aurora, CO 80045 USA
| |
Collapse
|
35
|
Ballesteros A, Swartz KJ. Regulation of membrane homeostasis by TMC1 mechanoelectrical transduction channels is essential for hearing. SCIENCE ADVANCES 2022; 8:eabm5550. [PMID: 35921424 PMCID: PMC9348795 DOI: 10.1126/sciadv.abm5550] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The mechanoelectrical transduction (MET) channel in auditory hair cells converts sound into electrical signals, enabling hearing. Transmembrane-like channel 1 and 2 (TMC1 and TMC2) are implicated in forming the pore of the MET channel. Here, we demonstrate that inhibition of MET channels, breakage of the tip links required for MET, or buffering of intracellular Ca... induces pronounced phosphatidylserine externalization, membrane blebbing, and ectosome release at the hair cell sensory organelle, culminating in the loss of TMC1. Membrane homeostasis triggered by MET channel inhibition requires Tmc1 but not Tmc2, and three deafness-causing mutations in Tmc1 cause constitutive phosphatidylserine externalization that correlates with deafness phenotype. Our results suggest that, in addition to forming the pore of the MET channel, TMC1 is a critical regulator of membrane homeostasis in hair cells, and that Tmc1-related hearing loss may involve alterations in membrane homeostasis.
Collapse
|
36
|
Recruitment of tetraspanin TSP-15 to epidermal wounds promotes plasma membrane repair in C. elegans. Dev Cell 2022; 57:1630-1642.e4. [PMID: 35777354 DOI: 10.1016/j.devcel.2022.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/26/2022] [Accepted: 06/07/2022] [Indexed: 01/14/2023]
Abstract
Maintaining the integrity of the plasma membrane after cellular damage is essential for cell survival. However, it is unclear how cells repair large membrane injuries in vivo. Here, we report that the tetraspanin protein, TSP-15, is recruited to large membrane wounds and forms a ring-like structure in C. elegans epidermis and promotes membrane repair after an injury. TSP-15 recruits from the adjacent region underneath the plasma membrane to the wound site in a RAB-5-dependent manner upon membrane damage. Genetic and live-imaging analysis suggested that the endosomal sorting complex required for transport III (ESCRT III) is necessary for recruiting TSP-15 from the early endosome to the damaged membrane. Moreover, TSP-15 interacts with and is required for the accumulation of t-SNARE protein Syntaxin-2, which facilitates membrane repair. These findings provide valuable insights into the role of the conserved tetraspanin TSP-15 in the cellular repair of large wounds resulting from environmental insults.
Collapse
|
37
|
Vasconcelos-Cardoso M, Batista-Almeida D, Rios-Barros LV, Castro-Gomes T, Girao H. Cellular and molecular mechanisms underlying plasma membrane functionality and integrity. J Cell Sci 2022; 135:275922. [PMID: 35801807 DOI: 10.1242/jcs.259806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The plasma membrane not only protects the cell from the extracellular environment, acting as a selective barrier, but also regulates cellular events that originate at the cell surface, playing a key role in various biological processes that are essential for the preservation of cell homeostasis. Therefore, elucidation of the mechanisms involved in the maintenance of plasma membrane integrity and functionality is of utmost importance. Cells have developed mechanisms to ensure the quality of proteins that inhabit the cell surface, as well as strategies to cope with injuries inflicted to the plasma membrane. Defects in these mechanisms can lead to the development or onset of several diseases. Despite the importance of these processes, a comprehensive and holistic perspective of plasma membrane quality control is still lacking. To tackle this gap, in this Review, we provide a thorough overview of the mechanisms underlying the identification and targeting of membrane proteins that are to be removed from the cell surface, as well as the membrane repair mechanisms triggered in both physiological and pathological conditions. A better understanding of the mechanisms underlying protein quality control at the plasma membrane can reveal promising and unanticipated targets for the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maria Vasconcelos-Cardoso
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Daniela Batista-Almeida
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Laura Valeria Rios-Barros
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Henrique Girao
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal.,Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| |
Collapse
|
38
|
Zou C, Zhang Y, Liu H, Wu Y, Zhou X. Extracellular Vesicles: Recent Insights Into the Interaction Between Host and Pathogenic Bacteria. Front Immunol 2022; 13:840550. [PMID: 35693784 PMCID: PMC9174424 DOI: 10.3389/fimmu.2022.840550] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are nanosized lipid particles released by virtually every living cell. EVs carry bioactive molecules, shuttle from cells to cells and transduce signals, regulating cell growth and metabolism. Pathogenic bacteria can cause serious infections via a wide range of strategies, and host immune systems also develop extremely complex adaptations to counteract bacterial infections. As notable carriers, EVs take part in the interaction between the host and bacteria in several approaches. For host cells, several strategies have been developed to resist bacteria via EVs, including expelling damaged membranes and bacteria, neutralizing toxins, triggering innate immune responses and provoking adaptive immune responses in nearly the whole body. For bacteria, EVs function as vehicles to deliver toxins and contribute to immune escape. Due to their crucial functions, EVs have great application potential in vaccines, diagnosis and treatments. In the present review, we highlight the most recent advances, application potential and remaining challenges in understanding EVs in the interaction between the host and bacteria.
Collapse
Affiliation(s)
- Chaoyu Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
- Department of Hematology and Hematology Research Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Yige Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yu Wu
- Department of Hematology and Hematology Research Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
39
|
Yan M, Xu M, Li Z, An Y, Wang Z, Li S, Chen Y, Xia Y, Wang L, Wang L, Ji S, Dong W, Shi J, Gao C. TMEM16F mediated phosphatidylserine exposure and microparticle release on erythrocyte contribute to hypercoagulable state in hyperuricemia. Blood Cells Mol Dis 2022; 96:102666. [PMID: 35567997 DOI: 10.1016/j.bcmd.2022.102666] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 12/15/2022]
Abstract
The link between hyperuricemia (HUA) and the risk of venous thromboembolism (VTE) has been well established. However, the mechanisms of thrombus generation and the effect of HUA on procoagulant activity (PCA) of erythrocytes remain unclear no matter in uremia or hyperuricemia. Here, phosphatidylserine (PS) exposure, microparticles (MPs) release, cytosolic Ca2+, TMEM16F expression, reactive oxygen species (ROS) and lipid peroxidation of erythrocyte were detected by flow cytometer. PCA was assessed by coagulation time, purified coagulation complex and fibrin production assays. The fibrin formation was observed by scanning electron microscopy (SEM). We found that PS exposure, MPs generation, TMEM16F expression and consequent PCA of erythrocyte in HUA patients significantly increased compared to those in healthy volunteers. Furthermore, high UA induced PS exposure, and MPs release of erythrocyte in concentration and time-dependent manners in vitro, which enhanced the PCA of erythrocyte and was inhibited by lactadherin, a PS inhibitor. Additionally, using SEM, we also observed compact fibrin clots with highly-branched networks and thin fibers supported by red blood cells (RBCs) and RBC-derived MPs (RMPs). Importantly, we demonstrated UA enhanced the production of ROS and lipid peroxidation and reduced the generation of glutathione (GSH) of erythrocyte, which enhanced TMEM16F activity and followed PS externalization and RMPs formation. Collectively, these results suggest that Ca2+-dependent TMEM16F activation may be responsible for UA-induced PS exposure and MPs release of RBC, which thereby contribute to the prothrombotic risk in HUA.
Collapse
Affiliation(s)
- Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Minghui Xu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Zhanni Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Zelong Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Shuli Li
- Department of Anesthesiology, Daqing Oil Field General Hospital, Daqing, China
| | - Yingli Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yanshi Xia
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Liqiu Wang
- Department of Clinical Laboratory, The Fifth Hospital, Harbin Medical University, Daqing, China
| | - Longlong Wang
- Department of Anesthesiology, Daqing Oil Field General Hospital, Daqing, China
| | - Shuting Ji
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Weijun Dong
- Department of General Surgery, The Fifth Hospital, Harbin Medical University, Daqing, China
| | - Jialan Shi
- Departments of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China.
| |
Collapse
|
40
|
Yamazaki Y, Kono K. Clathrin-mediated trafficking of phospholipid flippases is required for local plasma membrane/cell wall damage repair in budding yeast. Biochem Biophys Res Commun 2022; 606:156-162. [DOI: 10.1016/j.bbrc.2022.03.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022]
|
41
|
Ca 2+-activated sphingomyelin scrambling and turnover mediate ESCRT-independent lysosomal repair. Nat Commun 2022; 13:1875. [PMID: 35388011 PMCID: PMC8986845 DOI: 10.1038/s41467-022-29481-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/11/2022] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are vital organelles vulnerable to injuries from diverse materials. Failure to repair or sequester damaged lysosomes poses a threat to cell viability. Here we report that cells exploit a sphingomyelin-based lysosomal repair pathway that operates independently of ESCRT to reverse potentially lethal membrane damage. Various conditions perturbing organelle integrity trigger a rapid calcium-activated scrambling and cytosolic exposure of sphingomyelin. Subsequent metabolic conversion of sphingomyelin by neutral sphingomyelinases on the cytosolic surface of injured lysosomes promotes their repair, also when ESCRT function is compromised. Conversely, blocking turnover of cytosolic sphingomyelin renders cells more sensitive to lysosome-damaging drugs. Our data indicate that calcium-activated scramblases, sphingomyelin, and neutral sphingomyelinases are core components of a previously unrecognized membrane restoration pathway by which cells preserve the functional integrity of lysosomes.
Collapse
|
42
|
Kloc M, Uosef A, Wosik J, Kubiak JZ, Ghobrial RM. Virus interactions with the actin cytoskeleton-what we know and do not know about SARS-CoV-2. Arch Virol 2022; 167:737-749. [PMID: 35102456 PMCID: PMC8803281 DOI: 10.1007/s00705-022-05366-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
The actin cytoskeleton and actin-dependent molecular and cellular events are responsible for the organization of eukaryotic cells and their functions. Viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), depend on host cell organelles and molecular components for cell entry and propagation. Thus, it is not surprising that they also interact at many levels with the actin cytoskeleton of the host. There have been many studies on how different viruses reconfigure and manipulate the actin cytoskeleton of the host during successive steps of their life cycle. However, we know relatively little about the interactions of SARS-CoV-2 with the actin cytoskeleton. Here, we describe how the actin cytoskeleton is involved in the strategies used by different viruses for entry, assembly, and egress from the host cell. We emphasize what is known and unknown about SARS-CoV-2 in this regard. This review should encourage further investigation of the interactions of SARS-CoV-2 with cellular components, which will eventually be helpful for developing novel antiviral therapies for mitigating the severity of COVID-19.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA.
- Department of Genetics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Ahmed Uosef
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA
| | - Jarek Wosik
- Electrical and Computer Engineering Department, University of Houston, Houston, TX, 77204, USA
- Texas Center for Superconductivity, University of Houston, Houston, TX, 77204, USA
| | - Jacek Z Kubiak
- Military Institute of Medicine, Laboratory of Molecular Oncology and Innovative Therapies, Department of Oncology, 04-141, Warsaw, Poland
- Institute of Genetics and Development of Rennes, Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Univ. Rennes, UMR 6290, CNRS, 35000, Rennes, France
| | - Rafik M Ghobrial
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
43
|
Zhang F, Guo J, Zhang Z, Duan M, Wang G, Qian Y, Zhao H, Yang Z, Jiang X. Application of engineered extracellular vesicles for targeted tumor therapy. J Biomed Sci 2022; 29:14. [PMID: 35189894 PMCID: PMC8862579 DOI: 10.1186/s12929-022-00798-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
All cells, including prokaryotes and eukaryotes, could release extracellular vesicles (EVs). EVs contain many cellular components, including RNA, and surface proteins, and are essential for maintaining normal intercellular communication and homeostasis of the internal environment. EVs released from different tissues and cells exhibit excellent properties and functions (e.g., targeting specificity, regulatory ability, physical durability, and immunogenicity), rendering them a potential new option for drug delivery and precision therapy. EVs have been demonstrated to transport antitumor drugs for tumor therapy; additionally, EVs' contents and surface substance can be altered to improve their therapeutic efficacy in the clinic by boosting targeting potential and drug delivery effectiveness. EVs can regulate immune system function by affecting the tumor microenvironment, thereby inhibiting tumor progression. Co-delivery systems for EVs can be utilized to further improve the drug delivery efficiency of EVs, including hydrogels and liposomes. In this review, we discuss the isolation technologies of EVs, as well as engineering approaches to their modification. Moreover, we evaluate the therapeutic potential of EVs in tumors, including engineered extracellular vesicles and EVs' co-delivery systems. Technologies such as microfluidics can improve EVs isolation efficiency. Engineering technologies can improve EVs drug loading efficiency and tumor targeting. EVs-based drug co-delivery systems are being developed, such as those with liposomes and hydrogels.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinshuai Guo
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhenghou Zhang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Meiqi Duan
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Guang Wang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yiping Qian
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Haiying Zhao
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhi Yang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Xiaofeng Jiang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
44
|
Cheng Y, Feng S, Puchades C, Ko J, Figueroa E, Chen Y, Wu H, Gu S, Han T, Li J, Ho B, Shoichet B, Jan YN, Jan L. Identification of a conserved drug binding pocket in TMEM16 proteins. RESEARCH SQUARE 2022:rs.3.rs-1296933. [PMID: 35169791 PMCID: PMC8845511 DOI: 10.21203/rs.3.rs-1296933/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The TMEM16 family of calcium-activated membrane proteins includes ten mammalian paralogs (TMEM16A-K) playing distinct physiological roles with some implicated in cancer and airway diseases. Their modulators with therapeutic potential include 1PBC, a potent inhibitor with anti-tumoral properties, and the FDA-approved drug niclosamide that targets TMEM16F to inhibit syncytia formation induced by SARS-CoV-2 infection. Here, we report cryo-EM structures of TMEM16F associated with 1PBC and niclosamide, revealing that both molecules bind the same drug binding pocket. We functionally and computationally validate this binding pocket in TMEM16A as well as TMEM16F, thereby showing that drug modulation also involves residues that are not conserved between TMEM16A and TMEM16F. This study establishes a much-needed structural framework for the development of more potent and more specific drug molecules targeting TMEM16 proteins.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hao Wu
- University of California San Francisco
| | | | | | | | | | | | | | - Lily Jan
- University of California, San Francisco
| |
Collapse
|
45
|
Scramblases as Regulators of Proteolytic ADAM Function. MEMBRANES 2022; 12:membranes12020185. [PMID: 35207106 PMCID: PMC8880048 DOI: 10.3390/membranes12020185] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
Abstract
Proteolytic ectodomain release is a key mechanism for regulating the function of many cell surface proteins. The sheddases ADAM10 and ADAM17 are the best-characterized members of the family of transmembrane disintegrin-like metalloproteinase. Constitutive proteolytic activities are low but can be abruptly upregulated via inside-out signaling triggered by diverse activating events. Emerging evidence indicates that the plasma membrane itself must be assigned a dominant role in upregulation of sheddase function. Data are discussed that tentatively identify phospholipid scramblases as central players during these events. We propose that scramblase-dependent externalization of the negatively charged phospholipid phosphatidylserine (PS) plays an important role in the final activation step of ADAM10 and ADAM17. In this manuscript, we summarize the current knowledge on the interplay of cell membrane changes, PS exposure, and proteolytic activity of transmembrane proteases as well as the potential consequences in the context of immune response, infection, and cancer. The novel concept that scramblases regulate the action of ADAM-proteases may be extendable to other functional proteins that act at the cell surface.
Collapse
|
46
|
Le SC, Liang P, Lowry AJ, Yang H. Gating and Regulatory Mechanisms of TMEM16 Ion Channels and Scramblases. Front Physiol 2021; 12:787773. [PMID: 34867487 PMCID: PMC8640346 DOI: 10.3389/fphys.2021.787773] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
The transmembrane protein 16 (TMEM16) family consists of Ca2+-activated ion channels and Ca2+-activated phospholipid scramblases (CaPLSases) that passively flip-flop phospholipids between the two leaflets of the membrane bilayer. Owing to their diverse functions, TMEM16 proteins have been implicated in various human diseases, including asthma, cancer, bleeding disorders, muscular dystrophy, arthritis, epilepsy, dystonia, ataxia, and viral infection. To understand TMEM16 proteins in health and disease, it is critical to decipher their molecular mechanisms of activation gating and regulation. Structural, biophysical, and computational characterizations over the past decade have greatly advanced the molecular understanding of TMEM16 proteins. In this review, we summarize major structural features of the TMEM16 proteins with a focus on regulatory mechanisms and gating.
Collapse
Affiliation(s)
- Son C. Le
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
- Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
47
|
Proteomic analysis of necroptotic extracellular vesicles. Cell Death Dis 2021; 12:1059. [PMID: 34750357 PMCID: PMC8575773 DOI: 10.1038/s41419-021-04317-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022]
Abstract
Necroptosis is a regulated and inflammatory form of cell death. We, and others, have previously reported that necroptotic cells release extracellular vesicles (EVs). We have found that necroptotic EVs are loaded with proteins, including the phosphorylated form of the key necroptosis-executing factor, mixed lineage kinase domain-like kinase (MLKL). However, neither the exact protein composition, nor the impact, of necroptotic EVs have been delineated. To characterize their content, EVs from necroptotic and untreated U937 cells were isolated and analyzed by mass spectrometry-based proteomics. A total of 3337 proteins were identified, sharing a high degree of similarity with exosome proteome databases, and clearly distinguishing necroptotic and control EVs. A total of 352 proteins were significantly upregulated in the necroptotic EVs. Among these were MLKL and caspase-8, as validated by immunoblot. Components of the ESCRTIII machinery and inflammatory signaling were also upregulated in the necroptotic EVs, as well as currently unreported components of vesicle formation and transport, and necroptotic signaling pathways. Moreover, we found that necroptotic EVs can be phagocytosed by macrophages to modulate cytokine and chemokine secretion. Finally, we uncovered that necroptotic EVs contain tumor neoantigens, and are enriched with components of antigen processing and presentation. In summary, our study reveals a new layer of regulation during the early stage of necroptosis, mediated by the secretion of specific EVs that influences the microenvironment and may instigate innate and adaptive immune responses. This study sheds light on new potential players in necroptotic signaling and its related EVs, and uncovers the functional tasks accomplished by the cargo of these necroptotic EVs.
Collapse
|
48
|
Thiruvengadam G, Sreetama SC, Charton K, Hogarth M, Novak JS, Suel-Petat L, Chandra G, Allard B, Richard I, Jaiswal JK. Anoctamin 5 Knockout Mouse Model Recapitulates LGMD2L Muscle Pathology and Offers Insight Into in vivo Functional Deficits. J Neuromuscul Dis 2021; 8:S243-S255. [PMID: 34633328 PMCID: PMC8673513 DOI: 10.3233/jnd-210720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the Anoctamin 5 (Ano5) gene that result in the lack of expression or function of ANO5 protein, cause Limb Girdle Muscular Dystrophy (LGMD) 2L/R12, and Miyoshi Muscular Dystrophy (MMD3). However, the dystrophic phenotype observed in patient muscles is not uniformly recapitulated by ANO5 knockout in animal models of LGMD2L. Here we describe the generation of a mouse model of LGMD2L generated by targeted out-of-frame deletion of the Ano5 gene. This model shows progressive muscle loss, increased muscle weakness, and persistent bouts of myofiber regeneration without chronic muscle inflammation, which recapitulates the mild to moderate skeletal muscle dystrophy reported in the LGMD2L patients. We show that these features of ANO5 deficient muscle are not associated with a change in the calcium-activated sarcolemmal chloride channel activity or compromised in vivo regenerative myogenesis. Use of this mouse model allows conducting in vivo investigations into the functional role of ANO5 in muscle health and for preclinical therapeutic development for LGMD2L.
Collapse
Affiliation(s)
- Girija Thiruvengadam
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Sen Chandra Sreetama
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Karine Charton
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Marshall Hogarth
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - James S Novak
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| | - Laurence Suel-Petat
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Goutam Chandra
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGene, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Lyon, France
| | - Isabelle Richard
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jyoti K Jaiswal
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| |
Collapse
|
49
|
Chaudhary S, Patidar A, Dhiman A, Chaubey GK, Dilawari R, Talukdar S, Modanwal R, Raje M. Exposure of a specific pleioform of multifunctional glyceraldehyde 3-phosphate dehydrogenase initiates CD14-dependent clearance of apoptotic cells. Cell Death Dis 2021; 12:892. [PMID: 34593755 PMCID: PMC8482365 DOI: 10.1038/s41419-021-04168-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Rapid clearance of apoptotic cells by phagocytes is crucial for organogenesis, tissue homeostasis, and resolution of inflammation. This process is initiated by surface exposure of various 'eat me' ligands. Though phosphatidylserine (PS) is the best recognized general recognition ligand till date, recent studies have shown that PS by itself is not sufficient for clearance of apoptotic cells. In this study, we have identified a specific pleioform of GAPDH (Glyceraldehyde 3-phosphate dehydrogenase) that functions as an 'eat me' signal on apoptotic cell surface. This specific form of GAPDH which is exposed on surface of apoptotic cells was found to interact with CD14 present on plasma membrane of phagocytes leading to their engulfment. This is the first study demonstrating the novel interaction between multifunctional GAPDH and the phagocytic receptor CD14 resulting in apoptotic cell clearance (efferocytosis).
Collapse
Affiliation(s)
- Surbhi Chaudhary
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Anil Patidar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Asmita Dhiman
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | | | - Rahul Dilawari
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Sharmila Talukdar
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Radheshyam Modanwal
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India
| | - Manoj Raje
- Institute of Microbial Technology, CSIR, Sector 39A, Chandigarh, 160036, India.
| |
Collapse
|
50
|
Hu H, Liu M, Sun S. Pore-Forming Toxins During Bacterial Infection: Molecular Mechanisms and Potential Therapeutic Targets. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3773-3781. [PMID: 34522083 PMCID: PMC8434828 DOI: 10.2147/dddt.s322393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
Bacterial infections are predominantly treated with antibiotics, and resistance to antibiotics is becoming an increasing threat to our health. Pore-forming toxins (PFTs) are virulence factors secreted by many pathogenic bacterial strains, both in acute and chronic infections. They are special membrane-targeting proteins that exert toxic effects by forming pores in the cell membrane. Recent studies have elucidated the structure of PFTs and the detailed molecular mechanisms of their pathogenicity. Here, we discuss recent findings that highlight the regulatory mechanisms and important roles of two types of PFTs, α-PFTs and β-PFTs, in mediating the virulence of bacteria, and the therapeutic potential of targeting PFTs for antibacterial treatment. Therapeutic strategies based on PFTs are highly specific and may alleviate the issue of increasing resistance to antibiotics.
Collapse
Affiliation(s)
- Haijie Hu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - Min Liu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - Shuang Sun
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, People's Republic of China
| |
Collapse
|