1
|
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
|
2
|
Young LN, Sherrard A, Zhou H, Shaikh F, Hutchings J, Riggi M, Rosen MK, Giraldez AJ, Villa E. ExoSloNano: Multi-Modal Nanogold Tags for identification of Macromolecules in Live Cells & Cryo-Electron Tomograms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.617288. [PMID: 39416124 PMCID: PMC11482945 DOI: 10.1101/2024.10.12.617288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
In situ cryo-Electron Microscopy (cryo-EM) enables the direct interrogation of structure-function relationships by resolving macromolecular structures in their native cellular environment. Tremendous progress in sample preparation, imaging and data processing over the past decade has contributed to the identification and determination of large biomolecular complexes. However, the majority of proteins are of a size that still eludes identification in cellular cryo-EM data, and most proteins exist in low copy numbers. Therefore, novel tools are needed for cryo-EM to identify the vast majority of macromolecules across multiple size scales (from microns to nanometers). Here, we introduce and validate novel nanogold probes that enable the detection of specific proteins using cryo-ET (cryo-Electron Tomography) and resin-embedded correlated light and electron microscopy (CLEM). We demonstrate that these nanogold probes can be introduced into live cells, in a manner that preserves intact molecular networks and cell viability. We use this system to identify both cytoplasmic and nuclear proteins by room temperature EM, and resolve associated structures by cryo-ET. We further employ gold particles of different sizes to enable future multiplexed labeling and structural analysis. By providing high efficiency protein labeling in live cells and molecular specificity within cryo-ET tomograms, we establish a broadly enabling tool that significantly expands the proteome available to electron microscopy.
Collapse
Affiliation(s)
- Lindsey N Young
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | | | - Huabin Zhou
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Farhaz Shaikh
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Joshua Hutchings
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Margot Riggi
- Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Michael K Rosen
- University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, Dallas, TX, USA
| | | | - Elizabeth Villa
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Howard Hughes Medical Institute, La Jolla, CA, USA
| |
Collapse
|
3
|
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
|
4
|
Kachuei M, Zare R, Sayyahfar S, Khalili M, Movahedi H, Naghshbandi M, Eghdami S. An unusual imaging presentation of pediatric bacterial meningoencephalitis: a case-report study. Ann Med Surg (Lond) 2024; 86:1739-1744. [PMID: 38463079 PMCID: PMC10923334 DOI: 10.1097/ms9.0000000000001749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
Background Bacterial meningoencephalitis is a serious infection affecting the brain and its surrounding membranes. While imaging studies play a crucial role in diagnosing this condition, the typical radiological findings are well-documented. However, this case report describes an unusual imaging presentation that deviates from the expected patterns, emphasizing the need for awareness of such variations. Case presentation A 7-year-old female with no prior medical history was referred to our hospital with fever, seizure, and loss of Consciousness. She had mild flu a week before admission. The duration of seizure episodes were 2-3 min, with tonic-clonic uncontrollable jerky movements. Brudzinski and Kernig signs were positive and plantar reflex was upward bilaterally in the physical examination. The computed tomography (CT) scan showed brain ventriculomegaly/hydrocephalus, and MRI findings indicated multiple foci located at cerebellum, basal ganglia, and thalamus alongside intensely restricted diffusion of the layering debris, suggesting pyogenic ventriculitis. Cerebrospinal fluid (CSF) analysis showed severe hypoglycorrhachia, despite non-significant increase of protein. The patient was undergone antibiotic therapy with ceftriaxone, vancomycin and rifampin, resulting in normalization of CSF values. Conclusion This case report highlights the importance of recognizing and interpreting unusual imaging presentations of bacterial meningoencephalitis in paediatric patients. It emphasizes the need for a comprehensive diagnostic approach, including clinical evaluation, laboratory tests, and imaging studies, to ensure accurate diagnosis and appropriate management of this potentially life-threatening condition. Further research and awareness of atypical imaging findings are warranted to enhance our understanding and improve patient outcomes.
Collapse
Affiliation(s)
- Maryam Kachuei
- Firoozabadi Clinical Research Development Unit (FACRDU), School of Medicine, Iran University of Medical Sciences
| | - Ramin Zare
- Firoozabadi Clinical Research Development Unit (FACRDU), School of Medicine, Iran University of Medical Sciences
| | - Shirin Sayyahfar
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, School of Medicine, Iran University of Medical Sciences
| | - Mitra Khalili
- Department of Radiology, Shahid Beheshti University of Medical Sciences
| | | | | | - Shayan Eghdami
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Wang Z, Yang Y, Li S, Ma W, Wang K, Soberón M, Yan S, Shen J, Francis F, Bravo A, Zhang J. JAK/STAT signaling regulated intestinal regeneration defends insect pests against pore-forming toxins produced by Bacillus thuringiensis. PLoS Pathog 2024; 20:e1011823. [PMID: 38236820 PMCID: PMC10796011 DOI: 10.1371/journal.ppat.1011823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/13/2023] [Indexed: 01/22/2024] Open
Abstract
A variety of coordinated host-cell responses are activated as defense mechanisms against pore-forming toxins (PFTs). Bacillus thuringiensis (Bt) is a worldwide used biopesticide whose efficacy and precise application methods limits its use to replace synthetic pesticides in agricultural settings. Here, we analyzed the intestinal defense mechanisms of two lepidopteran insect pests after intoxication with sublethal dose of Bt PFTs to find out potential functional genes. We show that larval intestinal epithelium was initially damaged by the PFTs and that larval survival was observed after intestinal epithelium regeneration. Further analyses showed that the intestinal regeneration caused by Cry9A protein is regulated through c-Jun NH (2) terminal kinase (JNK) and Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathways. JAK/STAT signaling regulates intestinal regeneration through proliferation and differentiation of intestinal stem cells to defend three different Bt proteins including Cry9A, Cry1F or Vip3A in both insect pests, Chilo suppressalis and Spodoptera frugiperda. Consequently, a nano-biopesticide was designed to improve pesticidal efficacy based on the combination of Stat double stranded RNA (dsRNA)-nanoparticles and Bt strain. This formulation controlled insect pests with better effect suggesting its potential use to reduce the use of synthetic pesticides in agricultural settings for pest control.
Collapse
Affiliation(s)
- Zeyu Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanchao Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Sirui Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weihua Ma
- National Key Laboratory of Crop Genetic Improvement, National Centre of Plant Gene Research, Huazhong Agricultural University, Wuhan, China
| | - Kui Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Shuo Yan
- Department of Plant Biosecurity and MOA Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Plant Biosecurity and MOA Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Frederic Francis
- Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Jie Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
6
|
Bittel DC, Jaiswal JK. Early Endosomes Undergo Calcium-Triggered Exocytosis and Enable Repair of Diffuse and Focal Plasma Membrane Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300245. [PMID: 37705135 PMCID: PMC10667805 DOI: 10.1002/advs.202300245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/27/2023] [Indexed: 09/15/2023]
Abstract
Cells are routinely exposed to agents that cause plasma membrane (PM) injury. While pore-forming toxins (PFTs), and chemicals cause nanoscale holes dispersed throughout the PM, mechanical trauma causes focal lesions in the PM. To examine if these distinct injuries share common repair mechanism, membrane trafficking is monitored as the PM repairs from such injuries. During the course of repair, dispersed PM injury by the PFT Streptolysin O activates endocytosis, while focal mechanical injury to the PM inhibits endocytosis. Consequently, acute block of endocytosis prevents repair of diffuse, but not of focal injury. In contrast, a chronic block in endocytosis depletes cells of early endosomes and inhibits repair of focal injury. This study finds that both focal and diffuse PM injury activate Ca2+ -triggered exocytosis of early endosomes. The use of markers including endocytosed cargo, Rab5, Rab11, and VAMP3, all reveal injury-triggered exocytosis of early endosomes. Inhibiting Rab5 prevents injury-triggered early endosome exocytosis and phenocopies the failed PM repair of cells chronically depleted of early endosomes. These results identify early endosomes as a Ca2+ -regulated exocytic compartment, and uncover the requirement of their dual functions - endocytosis and regulated exocytosis, to differentially support PM repair based on the nature of the injury.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashington, DC20012USA
| |
Collapse
|
7
|
Chen F, Pang C, Zheng Z, Zhou W, Guo Z, Xiao D, Du H, Bravo A, Soberón M, Sun M, Peng D. Aminopeptidase MNP-1 triggers intestine protease production by activating daf-16 nuclear location to degrade pore-forming toxins in Caenorhabditis elegans. PLoS Pathog 2023; 19:e1011507. [PMID: 37440595 PMCID: PMC10368266 DOI: 10.1371/journal.ppat.1011507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Pore-forming toxins (PFTs) are effective tools for pathogens infection. By disrupting epithelial barriers and killing immune cells, PFTs promotes the colonization and reproduction of pathogenic microorganisms in their host. In turn, the host triggers defense responses, such as endocytosis, exocytosis, or autophagy. Bacillus thuringiensis (Bt) bacteria produce PFT, known as crystal proteins (Cry) which damage the intestinal cells of insects or nematodes, eventually killing them. In insects, aminopeptidase N (APN) has been shown to act as an important receptor for Cry toxins. Here, using the nematode Caenorhabditis elegans as model, an extensive screening of APN gene family was performed to analyze the potential role of these proteins in the mode of action of Cry5Ba against the nematode. We found that one APN, MNP-1, participate in the toxin defense response, since the mnp-1(ok2434) mutant showed a Cry5Ba hypersensitive phenotype. Gene expression analysis in mnp-1(ok2434) mutant revealed the involvement of two protease genes, F19C6.4 and R03G8.6, that participate in Cry5Ba degradation. Finally, analysis of the transduction pathway involved in F19C6.4 and R03G8.6 expression revealed that upon Cry5Ba exposure, the worms up regulated both protease genes through the activation of the FOXO transcription factor DAF-16, which was translocated into the nucleus. The nuclear location of DAF-16 was found to be dependent on mnp-1 under Cry5Ba treatment. Our work provides evidence of new host responses against PFTs produced by an enteric pathogenic bacterium, resulting in activation of host intestinal proteases that degrade the PFT in the intestine.
Collapse
Affiliation(s)
- Feng Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Cuiyun Pang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Ziqiang Zheng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Wei Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Zhiqing Guo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Danyang Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Hongwen Du
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
8
|
Bulgart HR, Goncalves I, Weisleder N. Leveraging Plasma Membrane Repair Therapeutics for Treating Neurodegenerative Diseases. Cells 2023; 12:1660. [PMID: 37371130 DOI: 10.3390/cells12121660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Plasma membrane repair is an essential cellular mechanism that reseals membrane disruptions after a variety of insults, and compromised repair capacity can contribute to the progression of many diseases. Neurodegenerative diseases are marked by membrane damage from many sources, reduced membrane integrity, elevated intracellular calcium concentrations, enhanced reactive oxygen species production, mitochondrial dysfunction, and widespread neuronal death. While the toxic intracellular effects of these changes in cellular physiology have been defined, the specific mechanism of neuronal death in certain neurodegenerative diseases remains unclear. An abundance of recent evidence indicates that neuronal membrane damage and pore formation in the membrane are key contributors to neurodegenerative disease pathogenesis. In this review, we have outlined evidence supporting the hypothesis that membrane damage is a contributor to neurodegenerative diseases and that therapeutically enhancing membrane repair can potentially combat neuronal death.
Collapse
Affiliation(s)
- Hannah R Bulgart
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Isabella Goncalves
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Noah Weisleder
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
9
|
Haram CS, Moitra S, Keane R, Kuhlmann FM, Frankfater C, Hsu FF, Beverley SM, Zhang K, Keyel PA. The sphingolipids ceramide and inositol phosphorylceramide protect the Leishmania major membrane from sterol-specific toxins. J Biol Chem 2023; 299:104745. [PMID: 37094699 PMCID: PMC10209034 DOI: 10.1016/j.jbc.2023.104745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023] Open
Abstract
The accessibility of sterols in mammalian cells to exogenous sterol-binding agents has been well-described previously, but sterol accessibility in distantly related protozoa is unclear. The human pathogen Leishmania major uses sterols and sphingolipids distinct from those used in mammals. Sterols in mammalian cells can be sheltered from sterol-binding agents by membrane components, including sphingolipids, but the surface exposure of ergosterol in Leishmania remains unknown. Here, we used flow cytometry to test the ability of the Leishmania major sphingolipids inositol phosphorylceramide (IPC), and ceramide to shelter ergosterol by preventing binding of the sterol-specific toxins streptolysin O and perfringolysin O and subsequent cytotoxicity. In contrast to mammalian systems, we found that Leishmania sphingolipids did not preclude toxin binding to sterols in the membrane. However, we show that IPC reduced cytotoxicity, and that ceramide reduced perfringolysin O-, but not streptolysin O-, mediated cytotoxicity in cells. Furthermore, we demonstrate ceramide sensing was controlled by the toxin L3 loop, and that ceramide was sufficient to protect L. major promastigotes from the anti-leishmaniasis drug amphotericin B. Based on these results, we propose a mechanism whereby pore-forming toxins engage additional lipids like ceramide to determine the optimal environment to sustain pore formation. Thus, L. major could serve as a genetically tractable protozoan model organism for understanding toxin-membrane interactions.
Collapse
Affiliation(s)
- Chaitanya S Haram
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409
| | - Samrat Moitra
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409
| | - Rilee Keane
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409
| | - F Matthew Kuhlmann
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Cheryl Frankfater
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Fong-Fu Hsu
- Mass Spectrometry Resource, Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen M Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409
| | - Peter A Keyel
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409.
| |
Collapse
|
10
|
Thapa R, Keyel PA. Patch repair protects cells from the small pore-forming toxin aerolysin. J Cell Sci 2023; 136:jcs261018. [PMID: 36951121 PMCID: PMC10198622 DOI: 10.1242/jcs.261018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
Aerolysin family pore-forming toxins damage the membrane, but membrane repair responses used to resist them, if any, remain controversial. Four proposed membrane repair mechanisms include toxin removal by caveolar endocytosis, clogging by annexins, microvesicle shedding catalyzed by MEK, and patch repair. Which repair mechanism aerolysin triggers is unknown. Membrane repair requires Ca2+, but it is controversial if Ca2+ flux is triggered by aerolysin. Here, we determined Ca2+ influx and repair mechanisms activated by aerolysin. In contrast to what is seen with cholesterol-dependent cytolysins (CDCs), removal of extracellular Ca2+ protected cells from aerolysin. Aerolysin triggered sustained Ca2+ influx. Intracellular Ca2+ chelation increased cell death, indicating that Ca2+-dependent repair pathways were triggered. Caveolar endocytosis failed to protect cells from aerolysin or CDCs. MEK-dependent repair did not protect against aerolysin. Aerolysin triggered slower annexin A6 membrane recruitment compared to CDCs. In contrast to what is seen with CDCs, expression of the patch repair protein dysferlin protected cells from aerolysin. We propose aerolysin triggers a Ca2+-dependent death mechanism that obscures repair, and the primary repair mechanism used to resist aerolysin is patch repair. We conclude that different classes of bacterial toxins trigger distinct repair mechanisms.
Collapse
Affiliation(s)
- Roshan Thapa
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Peter A. Keyel
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
11
|
Role of calcium-sensor proteins in cell membrane repair. Biosci Rep 2023; 43:232522. [PMID: 36728029 PMCID: PMC9970828 DOI: 10.1042/bsr20220765] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
Cell membrane repair is a critical process used to maintain cell integrity and survival from potentially lethal chemical, and mechanical membrane injury. Rapid increases in local calcium levels due to a membrane rupture have been widely accepted as a trigger for multiple membrane-resealing models that utilize exocytosis, endocytosis, patching, and shedding mechanisms. Calcium-sensor proteins, such as synaptotagmins (Syt), dysferlin, S100 proteins, and annexins, have all been identified to regulate, or participate in, multiple modes of membrane repair. Dysfunction of membrane repair from inefficiencies or genetic alterations in these proteins contributes to diseases such as muscular dystrophy (MD) and heart disease. The present review covers the role of some of the key calcium-sensor proteins and their involvement in membrane repair.
Collapse
|
12
|
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
|
13
|
Ariyama H. Visualizing the Domino-Like Prepore-to-Pore Transition of Streptolysin O by High-Speed AFM. J Membr Biol 2023; 256:91-103. [PMID: 35980453 PMCID: PMC9884259 DOI: 10.1007/s00232-022-00261-x] [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: 06/27/2022] [Accepted: 08/02/2022] [Indexed: 02/07/2023]
Abstract
Pore-forming proteins (PFPs) are produced by various organisms, including pathogenic bacteria, and form pores within the target cell membrane. Streptolysin O (SLO) is a PFP produced by Streptococcus pyogenes and forms high-order oligomers on the membrane surface. In this prepore state, multiple α-helices in domain 3 of each subunit exist as unfolded structures and transiently interact with each other. They subsequently transition into transmembrane β-hairpins (TMHs) and form pores with diameters of 20-30 nm. However, in this pore formation process, the trigger of the transition in a subunit and collaboration between subunits remains elusive. Here, I observed the dynamic pore formation process using high-speed atomic force microscopy. During the oligomer transition process, each subunit was sequentially inserted into the membrane, propagating along the oligomer in a domino-like fashion (chain reaction). This process also occurred on hybrid oligomers containing wildtype and mutant subunits, which cannot insert into the membrane because of an introduced disulfide bond. Furthermore, propagation still occurred when an excessive force was added to hybrid oligomers in the prepore state. Based on the observed chain reactions, I estimate the free energies and forces that trigger the transition in a subunit. Furthermore, I hypothesize that the collaboration between subunits is related to the structure of their TMH regions and interactions between TMH-TMH and TMH-lipid molecules.
Collapse
Affiliation(s)
- Hirotaka Ariyama
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192 Japan
| |
Collapse
|
14
|
Alves S, Pereira JM, Mayer RL, Gonçalves ADA, Impens F, Cabanes D, Sousa S. Cells Responding to Closely Related Cholesterol-Dependent Cytolysins Release Extracellular Vesicles with a Common Proteomic Content Including Membrane Repair Proteins. Toxins (Basel) 2022; 15:4. [PMID: 36668824 PMCID: PMC9865450 DOI: 10.3390/toxins15010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The plasma membrane (PM) protects cells from extracellular threats and supports cellular homeostasis. Some pathogens produce pore-forming toxins (PFTs) that disrupt PM integrity by forming transmembrane pores. High PFT concentrations cause massive damage leading to cell death and facilitating infection. Sub-lytic PFT doses activate repair mechanisms to restore PM integrity, support cell survival and limit disease. Shedding of extracellular vesicles (EVs) has been proposed as a key mechanism to eliminate PFT pores and restore PM integrity. We show here that cholesterol-dependent cytolysins (CDCs), a specific family of PFTs, are at least partially eliminated through EVs release, and we hypothesize that proteins important for PM repair might be included in EVs shed by cells during repair. To identify new PM repair proteins, we collected EVs released by cells challenged with sub-lytic doses of two different bacterial CDCs, listeriolysin O and pneumolysin, and determined the EV proteomic repertoire by LC-MS/MS. Intoxicated cells release similar EVs irrespectively of the CDC used. Also, they release more and larger EVs than non-intoxicated cells. A cluster of 70 proteins including calcium-binding proteins, molecular chaperones, cytoskeletal, scaffold and membrane trafficking proteins, was detected enriched in EVs collected from intoxicated cells. While some of these proteins have well-characterized roles in repair, the involvement of others requires further study. As proof of concept, we show here that Copine-1 and Copine-3, proteins abundantly detected in EVs released by intoxicated cells, are required for efficient repair of CDC-induced PM damage. Additionally, we reveal here new proteins potentially involved in PM repair and give new insights into common mechanisms and machinery engaged by cells in response to PM damage.
Collapse
Affiliation(s)
- Sara Alves
- Cell Biology of Bacterial Infections, IBMC, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana M. Pereira
- Cell Biology of Bacterial Infections, IBMC, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Molecular and Cellular (MC) Biology PhD Program, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Rupert L. Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB Proteomics Core, VIB, 9052 Ghent, Belgium
| | - Alexandre D. A. Gonçalves
- Cell Biology of Bacterial Infections, IBMC, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB Proteomics Core, VIB, 9052 Ghent, Belgium
| | - Didier Cabanes
- Molecular Microbiology, IBMC, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Sandra Sousa
- Cell Biology of Bacterial Infections, IBMC, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
15
|
Haram CS, Moitra S, Keane R, Breslav E, Zhang K, Keyel PA. Deciphering the Molecular Mechanism and Function of Pore-Forming Toxins using Leishmania major. J Vis Exp 2022:10.3791/64341. [PMID: 36373947 PMCID: PMC9792151 DOI: 10.3791/64341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2023] Open
Abstract
Understanding the function and mechanism of pore-forming toxins (PFTs) is challenging because cells resist the membrane damage caused by PFTs. While biophysical approaches help understand pore formation, they often rely on reductionist approaches lacking the full complement of membrane lipids and proteins. Cultured human cells provide an alternative system, but their complexity and redundancies in repair mechanisms make identifying specific mechanisms difficult. In contrast, the human protozoan pathogen responsible for cutaneous leishmaniasis, Leishmania major, offers an optimal balance between complexity and physiologic relevance. L. major is genetically tractable and can be cultured to high density in vitro, and any impact of perturbations on infection can be measured in established murine models. In addition, L. major synthesizes lipids distinct from their mammalian counterparts, which could alter membrane dynamics. These alterations in membrane dynamics can be probed with PFTs from the best-characterized toxin family, cholesterol-dependent cytolysins (CDCs). CDCs bind to ergosterol in the Leishmania membrane and can kill L. major promastigotes, indicating that L. major is a suitable model system for determining the cellular and molecular mechanisms of PFT function. This work describes methods for testing PFT function in L. major promastigotes, including parasite culture, genetic tools for assessing lipid susceptibility, membrane binding assays, and cell death assays. These assays will enable the rapid use of L. major as a powerful model system for understanding PFT function across a range of evolutionarily diverse organisms and commonalities in lipid organization.
Collapse
Affiliation(s)
| | - Samrat Moitra
- Department of Biological Sciences, Texas Tech University
| | - Rilee Keane
- Department of Biological Sciences, Texas Tech University
| | - Elana Breslav
- Department of Biological Sciences, Texas Tech University
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University
| | - Peter A Keyel
- Department of Biological Sciences, Texas Tech University;
| |
Collapse
|
16
|
Hui J, Stjepić V, Nakamura M, Parkhurst SM. Wrangling Actin Assemblies: Actin Ring Dynamics during Cell Wound Repair. Cells 2022; 11:2777. [PMID: 36139352 PMCID: PMC9497110 DOI: 10.3390/cells11182777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 12/18/2022] Open
Abstract
To cope with continuous physiological and environmental stresses, cells of all sizes require an effective wound repair process to seal breaches to their cortex. Once a wound is recognized, the cell must rapidly plug the injury site, reorganize the cytoskeleton and the membrane to pull the wound closed, and finally remodel the cortex to return to homeostasis. Complementary studies using various model organisms have demonstrated the importance and complexity behind the formation and translocation of an actin ring at the wound periphery during the repair process. Proteins such as actin nucleators, actin bundling factors, actin-plasma membrane anchors, and disassembly factors are needed to regulate actin ring dynamics spatially and temporally. Notably, Rho family GTPases have been implicated throughout the repair process, whereas other proteins are required during specific phases. Interestingly, although different models share a similar set of recruited proteins, the way in which they use them to pull the wound closed can differ. Here, we describe what is currently known about the formation, translocation, and remodeling of the actin ring during the cell wound repair process in model organisms, as well as the overall impact of cell wound repair on daily events and its importance to our understanding of certain diseases and the development of therapeutic delivery modalities.
Collapse
Affiliation(s)
| | | | | | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Souissi W, Alistair T, Domanska B, Fortea E, West MJ, Schwartz JL, Crickmore N. Probing the Mechanism of Action of Cry41Aa on HepG2 through the Establishment of a Resistant Subline. Toxins (Basel) 2022; 14:319. [PMID: 35622566 PMCID: PMC9147150 DOI: 10.3390/toxins14050319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Cry41Aa, also called parasporin-3, belongs to a group of toxins from the entomopathogenic bacterium Bacillus thuringiensis that show activity against human cancer cells. Cry41Aa exhibits preferential cytocidal activity towards HL-60 (human promyelocytic leukaemia cells) and HepG2 (human liver cancer cells) cell lines after being proteolytically activated. To better understand the mechanism of action of Cry41Aa, we evolved resistance in HepG2 cells through repeated exposure to increasing doses of the toxin. Concentrations of Cry41Aa that killed over 50% of the parental HepG2 cells had no significant effect on the viability of the resistant cells and did not induce either pore formation or p38 phosphorylation (both characteristic features of pore-forming toxins). Preliminary RNA sequencing data identified AQP9 as a potential mediator of resistance, but extensive investigations failed to show a causal link and did not support an enhanced cell repair process as the resistance mechanism.
Collapse
Affiliation(s)
- Wided Souissi
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (T.A.); (B.D.); (M.J.W.); (N.C.)
| | - Tweedie Alistair
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (T.A.); (B.D.); (M.J.W.); (N.C.)
| | - Barbara Domanska
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (T.A.); (B.D.); (M.J.W.); (N.C.)
| | - Eva Fortea
- Departement of Pharmacology et Physiology, University of Montreal, Montreal, QC H3C 3J7, Canada; (E.F.); (J.-L.S.)
| | - Michelle J. West
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (T.A.); (B.D.); (M.J.W.); (N.C.)
| | - Jean-Louis Schwartz
- Departement of Pharmacology et Physiology, University of Montreal, Montreal, QC H3C 3J7, Canada; (E.F.); (J.-L.S.)
| | - Neil Crickmore
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK; (T.A.); (B.D.); (M.J.W.); (N.C.)
| |
Collapse
|
18
|
Paleo BJ, McElhanon KE, Bulgart HR, Banford KK, Beck EX, Sattler KM, Goines BN, Ratcliff SL, Crowe KE, Weisleder N. Reduced Sarcolemmal Membrane Repair Exacerbates Striated Muscle Pathology in a Mouse Model of Duchenne Muscular Dystrophy. Cells 2022; 11:1417. [PMID: 35563723 PMCID: PMC9100510 DOI: 10.3390/cells11091417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a common X-linked degenerative muscle disorder that involves mutations in the DMD gene that frequently reduce the expression of the dystrophin protein, compromising the structural integrity of the sarcolemmal membrane and leaving it vulnerable to injury during cycles of muscle contraction and relaxation. This results in an increased frequency of sarcolemma disruptions that can compromise the barrier function of the membrane and lead to death of the myocyte. Sarcolemmal membrane repair processes can potentially compensate for increased membrane disruptions in DMD myocytes. Previous studies demonstrated that TRIM72, a muscle-enriched tripartite motif (TRIM) family protein also known as mitsugumin 53 (MG53), is a component of the cell membrane repair machinery in striated muscle. To test the importance of membrane repair in striated muscle in compensating for the membrane fragility in DMD, we crossed TRIM72/MG53 knockout mice into the mdx mouse model of DMD. These double knockout (DKO) mice showed compromised sarcolemmal membrane integrity compared to mdx mice, as measured by immunoglobulin G staining and ex vivo muscle laser microscopy wounding assays. We also found a significant decrease in muscle ex vivo contractile function as compared to mdx mice at both 6 weeks and 1.5 years of age. As the DKO mice aged, they developed more extensive fibrosis in skeletal muscles compared to mdx. Our findings indicate that TRIM72/MG53-mediated membrane repair can partially compensate for the sarcolemmal fragility associated with DMD and that the loss of membrane repair results in increased pathology in the DKO mice.
Collapse
Affiliation(s)
- Brian J. Paleo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kevin E. McElhanon
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Hannah R. Bulgart
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kassidy K. Banford
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Eric X Beck
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| | - Kristina M. Sattler
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Briana N. Goines
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Shelby L. Ratcliff
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Kelly E. Crowe
- Department of Biology, School of Behavioral & Natural Sciences, Mount St. Joseph University, Cincinnati, OH 45233, USA; (K.M.S.); (B.N.G.); (S.L.R.); (K.E.C.)
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; (B.J.P.); (K.E.M.); (H.R.B.); (K.K.B.); (E.X.B.)
| |
Collapse
|
19
|
Ormsby TJR, Owens SE, Clement L, Mills TJ, Cronin JG, Bromfield JJ, Sheldon IM. Oxysterols Protect Epithelial Cells Against Pore-Forming Toxins. Front Immunol 2022; 13:815775. [PMID: 35154132 PMCID: PMC8825411 DOI: 10.3389/fimmu.2022.815775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/05/2022] [Indexed: 12/25/2022] Open
Abstract
Many species of bacteria produce toxins such as cholesterol-dependent cytolysins that form pores in cell membranes. Membrane pores facilitate infection by releasing nutrients, delivering virulence factors, and causing lytic cell damage - cytolysis. Oxysterols are oxidized forms of cholesterol that regulate cellular cholesterol and alter immune responses to bacteria. Whether oxysterols also influence the protection of cells against pore-forming toxins is unresolved. Here we tested the hypothesis that oxysterols stimulate the intrinsic protection of epithelial cells against damage caused by cholesterol-dependent cytolysins. We treated epithelial cells with oxysterols and then challenged them with the cholesterol-dependent cytolysin, pyolysin. Treating HeLa cells with 27-hydroxycholesterol, 25-hydroxycholesterol, 7α-hydroxycholesterol, or 7β-hydroxycholesterol reduced pyolysin-induced leakage of lactate dehydrogenase and reduced pyolysin-induced cytolysis. Specifically, treatment with 10 ng/ml 27-hydroxycholesterol for 24 h reduced pyolysin-induced lactate dehydrogenase leakage by 88%, and reduced cytolysis from 74% to 1%. Treating HeLa cells with 27-hydroxycholesterol also reduced pyolysin-induced leakage of potassium ions, prevented mitogen-activated protein kinase cell stress responses, and limited alterations in the cytoskeleton. Furthermore, 27-hydroxycholesterol reduced pyolysin-induced damage in lung and liver epithelial cells, and protected against the cytolysins streptolysin O and Staphylococcus aureus α-hemolysin. Although oxysterols regulate cellular cholesterol by activating liver X receptors, cytoprotection did not depend on liver X receptors or changes in total cellular cholesterol. However, oxysterol cytoprotection was partially dependent on acyl-CoA:cholesterol acyltransferase (ACAT) reducing accessible cholesterol in cell membranes. Collectively, these findings imply that oxysterols stimulate the intrinsic protection of epithelial cells against pore-forming toxins and may help protect tissues against pathogenic bacteria.
Collapse
Affiliation(s)
- Thomas J R Ormsby
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Sian E Owens
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Liam Clement
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Tom J Mills
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - James G Cronin
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - John J Bromfield
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Iain Martin Sheldon
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| |
Collapse
|
20
|
Lata K, Singh M, Chatterjee S, Chattopadhyay K. Membrane Dynamics and Remodelling in Response to the Action of the Membrane-Damaging Pore-Forming Toxins. J Membr Biol 2022; 255:161-173. [PMID: 35305136 DOI: 10.1007/s00232-022-00227-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
Abstract
Pore-forming protein toxins (PFTs) represent a diverse class of membrane-damaging proteins that are produced by a wide variety of organisms. PFT-mediated membrane perforation is largely governed by the chemical composition and the physical properties of the plasma membranes. The interaction between the PFTs with the target membranes is critical for the initiation of the pore-formation process, and can lead to discrete membrane reorganization events that further aids in the process of pore-formation. Punching holes on the plasma membranes by the PFTs interferes with the cellular homeostasis by disrupting the ion-balance inside the cells that in turn can turn on multiple signalling cascades required to restore membrane integrity and cellular homeostasis. In this review, we discuss the physicochemical attributes of the plasma membranes associated with the pore-formation processes by the PFTs, and the subsequent membrane remodelling events that may start off the membrane-repair mechanisms.
Collapse
Affiliation(s)
- Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Mahendra Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Shamaita Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India.
| |
Collapse
|
21
|
Ray S, Roth R, Keyel PA. Membrane repair triggered by cholesterol-dependent cytolysins is activated by mixed lineage kinases and MEK. SCIENCE ADVANCES 2022; 8:eabl6367. [PMID: 35294243 PMCID: PMC8926344 DOI: 10.1126/sciadv.abl6367] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Repair of plasma membranes damaged by bacterial pore-forming toxins, such as streptolysin O or perfringolysin O, during septic cardiomyopathy or necrotizing soft tissue infections is mediated by several protein families. However, the activation of these proteins downstream of ion influx is poorly understood. Here, we demonstrate that following membrane perforation by bacterial cholesterol-dependent cytolysins, calcium influx activates mixed lineage kinase 3 independently of protein kinase C or ceramide generation. Mixed lineage kinase 3 uncouples mitogen-activated kinase kinase (MEK) and extracellular-regulated kinase (ERK) signaling. MEK signals via an ERK-independent pathway to promote rapid annexin A2 membrane recruitment and enhance microvesicle shedding. This pathway accounted for 70% of all calcium ion-dependent repair responses to streptolysin O and perfringolysin O, but only 50% of repair to intermedilysin. We conclude that mixed lineage kinase signaling via MEK coordinates microvesicle shedding, which is critical for cellular survival against cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
- Sucharit Ray
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Robyn Roth
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Peter A. Keyel
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Corresponding author.
| |
Collapse
|
22
|
Westman J, Plumb J, Licht A, Yang M, Allert S, Naglik JR, Hube B, Grinstein S, Maxson ME. Calcium-dependent ESCRT recruitment and lysosome exocytosis maintain epithelial integrity during Candida albicans invasion. Cell Rep 2022; 38:110187. [PMID: 34986345 PMCID: PMC8755444 DOI: 10.1016/j.celrep.2021.110187] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/21/2021] [Accepted: 12/07/2021] [Indexed: 01/12/2023] Open
Abstract
Candida albicans is both a commensal and an opportunistic fungal pathogen. Invading hyphae of C. albicans secrete candidalysin, a pore-forming peptide toxin. To prevent cell death, epithelial cells must protect themselves from direct damage induced by candidalysin and by the mechanical forces exerted by expanding hyphae. We identify two key Ca2+-dependent repair mechanisms employed by epithelial cells to withstand candidalysin-producing hyphae. Using camelid nanobodies, we demonstrate candidalysin secretion directly into the invasion pockets induced by elongating C. albicans hyphae. The toxin induces oscillatory increases in cytosolic [Ca2+], which cause hydrolysis of PtdIns(4,5)P2 and loss of cortical actin. Epithelial cells dispose of damaged membrane regions containing candidalysin by an Alg-2/Alix/ESCRT-III-dependent blebbing process. At later stages, plasmalemmal tears induced mechanically by invading hyphae are repaired by exocytic insertion of lysosomal membranes. These two repair mechanisms maintain epithelial integrity and prevent mucosal damage during both commensal growth and infection by C. albicans.
Collapse
Affiliation(s)
- Johannes Westman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jonathan Plumb
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Anna Licht
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Mabel Yang
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), 07745 Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), 07745 Jena, Germany; Institute of Microbiology, Friedrich Schiller University, 07745 Jena, Germany.
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5C 1N8, Canada.
| | - Michelle E Maxson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
23
|
Bittel DC, Sreetama SC, Chandra G, Ziegler R, Nagaraju K, Van der Meulen JH, Jaiswal JK. Secreted acid sphingomyelinase as a potential gene therapy for limb girdle muscular dystrophy 2B. J Clin Invest 2022; 132:e141295. [PMID: 34981776 PMCID: PMC8718136 DOI: 10.1172/jci141295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Efficient sarcolemmal repair is required for muscle cell survival, with deficits in this process leading to muscle degeneration. Lack of the sarcolemmal protein dysferlin impairs sarcolemmal repair by reducing secretion of the enzyme acid sphingomyelinase (ASM), and causes limb girdle muscular dystrophy 2B (LGMD2B). The large size of the dysferlin gene poses a challenge for LGMD2B gene therapy efforts aimed at restoring dysferlin expression in skeletal muscle fibers. Here, we present an alternative gene therapy approach targeting reduced ASM secretion, the consequence of dysferlin deficit. We showed that the bulk endocytic ability is compromised in LGMD2B patient cells, which was addressed by extracellularly treating cells with ASM. Expression of secreted human ASM (hASM) using a liver-specific adeno-associated virus (AAV) vector restored membrane repair capacity of patient cells to healthy levels. A single in vivo dose of hASM-AAV in the LGMD2B mouse model restored myofiber repair capacity, enabling efficient recovery of myofibers from focal or lengthening contraction-induced injury. hASM-AAV treatment was safe, attenuated fibro-fatty muscle degeneration, increased myofiber size, and restored muscle strength, similar to dysferlin gene therapy. These findings elucidate the role of ASM in dysferlin-mediated plasma membrane repair and to our knowledge offer the first non-muscle-targeted gene therapy for LGMD2B.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Robin Ziegler
- Rare and Neurologic Diseases Research, Sanofi, Framingham, Massachusetts, USA
| | - Kanneboyina Nagaraju
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, New York, USA
| | | | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
24
|
Actin Cytoskeletal Dynamics in Single-Cell Wound Repair. Int J Mol Sci 2021; 22:ijms221910886. [PMID: 34639226 PMCID: PMC8509258 DOI: 10.3390/ijms221910886] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
The plasma membrane protects the eukaryotic cell from its surroundings and is essential for cell viability; thus, it is crucial that membrane disruptions are repaired quickly to prevent immediate dyshomeostasis and cell death. Accordingly, cells have developed efficient repair mechanisms to rapidly reseal ruptures and reestablish membrane integrity. The cortical actin cytoskeleton plays an instrumental role in both plasma membrane resealing and restructuring in response to damage. Actin directly aids membrane repair or indirectly assists auxiliary repair mechanisms. Studies investigating single-cell wound repair have often focused on the recruitment and activation of specialized repair machinery, despite the undeniable need for rapid and dynamic cortical actin modulation; thus, the role of the cortical actin cytoskeleton during wound repair has received limited attention. This review aims to provide a comprehensive overview of membrane repair mechanisms directly or indirectly involving cortical actin cytoskeletal remodeling.
Collapse
|
25
|
Yang Y, Chen Y, Guo J, Liu H, Ju H. A pore-forming protein-induced surface-enhanced Raman spectroscopic strategy for dynamic tracing of cell membrane repair. iScience 2021; 24:102980. [PMID: 34485862 PMCID: PMC8403736 DOI: 10.1016/j.isci.2021.102980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/10/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
The plasma membrane repair holds significance for maintaining cell survival and homeostasis. To achieve the sensitive visualization of membrane repair process for revealing its mechanism, this work designs a perforation-induced surface-enhanced Raman spectroscopy (SERS) strategy by conjugating Raman reporter (4-mercaptobenzoic acid) loaded gold nanostars with pore-forming protein streptolysin O (SLO) to induce the SERS signal on living cells. The SERS signal obviously decreases with the initiation of membrane repair and the degradation of SLO pores due to the departure of gold-nanostar-conjugated SLO. Thus, the designed strategy can dynamically visualize the complete cell membrane repair and provide a sensitive method to demonstrate the SLO endocytosis- and exocytosis-mediated repairing mechanism. Using DOX-resistant MCF-7 cells as a model, a timely repair-blocking technology for promoting the highly efficient treatment of drug-resistant cancer cells is also proposed. This work opens an avenue for probing the plasma membrane repairing mechanisms and designing the precision therapeutic schedule.
Collapse
Affiliation(s)
- Yuanjiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P.R. China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P.R. China
| | - Jingxing Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P.R. China
| | - Huipu Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P.R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P.R. China
| |
Collapse
|
26
|
Ilangumaran Ponmalar I, Sarangi NK, Basu JK, Ayappa KG. Pore Forming Protein Induced Biomembrane Reorganization and Dynamics: A Focused Review. Front Mol Biosci 2021; 8:737561. [PMID: 34568431 PMCID: PMC8459938 DOI: 10.3389/fmolb.2021.737561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Pore forming proteins are a broad class of pathogenic proteins secreted by organisms as virulence factors due to their ability to form pores on the target cell membrane. Bacterial pore forming toxins (PFTs) belong to a subclass of pore forming proteins widely implicated in bacterial infections. Although the action of PFTs on target cells have been widely investigated, the underlying membrane response of lipids during membrane binding and pore formation has received less attention. With the advent of superresolution microscopy as well as the ability to carry out molecular dynamics (MD) simulations of the large protein membrane assemblies, novel microscopic insights on the pore forming mechanism have emerged over the last decade. In this review, we focus primarily on results collated in our laboratory which probe dynamic lipid reorganization induced in the plasma membrane during various stages of pore formation by two archetypal bacterial PFTs, cytolysin A (ClyA), an α-toxin and listeriolysin O (LLO), a β-toxin. The extent of lipid perturbation is dependent on both the secondary structure of the membrane inserted motifs of pore complex as well as the topological variations of the pore complex. Using confocal and superresolution stimulated emission depletion (STED) fluorescence correlation spectroscopy (FCS) and MD simulations, lipid diffusion, cholesterol reorganization and deviations from Brownian diffusion are correlated with the oligomeric state of the membrane bound protein as well as the underlying membrane composition. Deviations from free diffusion are typically observed at length scales below ∼130 nm to reveal the presence of local dynamical heterogeneities that emerge at the nanoscale-driven in part by preferential protein binding to cholesterol and domains present in the lipid membrane. Interrogating the lipid dynamics at the nanoscale allows us further differentiate between binding and pore formation of β- and α-PFTs to specific domains in the membrane. The molecular insights gained from the intricate coupling that occurs between proteins and membrane lipids and receptors during pore formation are expected to improve our understanding of the virulent action of PFTs.
Collapse
Affiliation(s)
| | - Nirod K. Sarangi
- School of Chemical Science, Dublin City University, Dublin, Ireland
| | - Jaydeep K. Basu
- Department of Physics, Indian Institute of Science, Bangalore, India
| | - K. Ganapathy Ayappa
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
27
|
Kulma M, Anderluh G. Beyond pore formation: reorganization of the plasma membrane induced by pore-forming proteins. Cell Mol Life Sci 2021; 78:6229-6249. [PMID: 34387717 PMCID: PMC11073440 DOI: 10.1007/s00018-021-03914-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 12/23/2022]
Abstract
Pore-forming proteins (PFPs) are a heterogeneous group of proteins that are expressed and secreted by a wide range of organisms. PFPs are produced as soluble monomers that bind to a receptor molecule in the host cell membrane. They then assemble into oligomers that are incorporated into the lipid membrane to form transmembrane pores. Such pore formation alters the permeability of the plasma membrane and is one of the most common mechanisms used by PFPs to destroy target cells. Interestingly, PFPs can also indirectly manipulate diverse cellular functions. In recent years, increasing evidence indicates that the interaction of PFPs with lipid membranes is not only limited to pore-induced membrane permeabilization but is also strongly associated with extensive plasma membrane reorganization. This includes lateral rearrangement and deformation of the lipid membrane, which can lead to the disruption of target cell function and finally death. Conversely, these modifications also constitute an essential component of the membrane repair system that protects cells from the lethal consequences of pore formation. Here, we provide an overview of the current knowledge on the changes in lipid membrane organization caused by PFPs from different organisms.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1001, Ljubljana, Slovenia
| |
Collapse
|
28
|
Claude-Taupin A, Jia J, Bhujabal Z, Garfa-Traoré M, Kumar S, da Silva GPD, Javed R, Gu Y, Allers L, Peters R, Wang F, da Costa LJ, Pallikkuth S, Lidke KA, Mauthe M, Verlhac P, Uchiyama Y, Salemi M, Phinney B, Tooze SA, Mari MC, Johansen T, Reggiori F, Deretic V. ATG9A protects the plasma membrane from programmed and incidental permeabilization. Nat Cell Biol 2021; 23:846-858. [PMID: 34257406 PMCID: PMC8276549 DOI: 10.1038/s41556-021-00706-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
The integral membrane protein ATG9A plays a key role in autophagy. It displays a broad intracellular distribution and is present in numerous compartments, including the plasma membrane (PM). The reasons for the distribution of ATG9A to the PM and its role at the PM are not understood. Here, we show that ATG9A organizes, in concert with IQGAP1, components of the ESCRT system and uncover cooperation between ATG9A, IQGAP1 and ESCRTs in protection from PM damage. ESCRTs and ATG9A phenocopied each other in protection against PM injury. ATG9A knockouts sensitized the PM to permeabilization by a broad spectrum of microbial and endogenous agents, including gasdermin, MLKL and the MLKL-like action of coronavirus ORF3a. Thus, ATG9A engages IQGAP1 and the ESCRT system to maintain PM integrity.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Zambarlal Bhujabal
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Meriem Garfa-Traoré
- Cell Imaging Platform, INSERM US24 Structure Fédérative de Recherche Necker, Université de Paris, Paris, France
| | - Suresh Kumar
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Gustavo Peixoto Duarte da Silva
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yuexi Gu
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ryan Peters
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Fulong Wang
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Luciana Jesus da Costa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandeep Pallikkuth
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - Mario Mauthe
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pauline Verlhac
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Sharon A Tooze
- The Francis Crick Institute, Molecular Cell Biology of Autophagy Laboratory, London, UK
| | - Muriel C Mari
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolic (AIM) Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
29
|
Pospiech M, Owens SE, Miller DJ, Austin-Muttitt K, Mullins JGL, Cronin JG, Allemann RK, Sheldon IM. Bisphosphonate inhibitors of squalene synthase protect cells against cholesterol-dependent cytolysins. FASEB J 2021; 35:e21640. [PMID: 33991130 DOI: 10.1096/fj.202100164r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 01/29/2023]
Abstract
Certain species of pathogenic bacteria damage tissues by secreting cholesterol-dependent cytolysins, which form pores in the plasma membranes of animal cells. However, reducing cholesterol protects cells against these cytolysins. As the first committed step of cholesterol biosynthesis is catalyzed by squalene synthase, we explored whether inhibiting this enzyme protected cells against cholesterol-dependent cytolysins. We first synthesized 22 different nitrogen-containing bisphosphonate molecules that were designed to inhibit squalene synthase. Squalene synthase inhibition was quantified using a cell-free enzyme assay, and validated by computer modeling of bisphosphonate molecules binding to squalene synthase. The bisphosphonates were then screened for their ability to protect HeLa cells against the damage caused by the cholesterol-dependent cytolysin, pyolysin. The most effective bisphosphonate reduced pyolysin-induced leakage of lactate dehydrogenase into cell supernatants by >80%, and reduced pyolysin-induced cytolysis from >75% to <25%. In addition, this bisphosphonate reduced pyolysin-induced leakage of potassium from cells, limited changes in the cytoskeleton, prevented mitogen-activated protein kinases cell stress responses, and reduced cellular cholesterol. The bisphosphonate also protected cells against another cholesterol-dependent cytolysin, streptolysin O, and protected lung epithelial cells and primary dermal fibroblasts against cytolysis. Our findings imply that treatment with bisphosphonates that inhibit squalene synthase might help protect tissues against pathogenic bacteria that secrete cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
- Mateusz Pospiech
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Siân E Owens
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | | | | | - James G Cronin
- Swansea University Medical School, Swansea University, Swansea, UK
| | | | - I Martin Sheldon
- Swansea University Medical School, Swansea University, Swansea, UK
| |
Collapse
|
30
|
Ilangumaran Ponmalar I, Ayappa KG, Basu JK. Bacterial protein listeriolysin O induces nonmonotonic dynamics because of lipid ejection and crowding. Biophys J 2021; 120:3040-3049. [PMID: 34214525 DOI: 10.1016/j.bpj.2021.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 01/15/2023] Open
Abstract
Membrane-bound protein complexes involving pore forming toxins (PFTs) released by virulent bacteria are known to form transmembrane pores leading to host cell lysis. Developing alternative strategies against PFT mediated bacterial virulence factors requires an understanding of the cellular membrane response. However, membrane disruption and related lipid reorganization events during attack by PFTs remain largely unexplored. We report counterintuitive and nonmonotonic variations in lipid diffusion, measured using confocal fluorescence correlation spectroscopy, due to interplay of lipid ejection and crowding by membrane-bound oligomers of a prototypical cholesterol-dependent cytolysin, listeriolysin O (LLO). The observed dynamical crossover is correlated with concentration dependent transitions of LLO oligomeric state populations from rings to arc-like pore complexes, predicted using a proposed two-state free area-based diffusion model. At low PFT concentrations, a hitherto unexplored regime of increased lipid diffusivity is attributed to lipid ejection events because of a preponderance of ring-like pore states. At higher protein concentrations in which membrane-inserted arc-like pores dominate, lipid ejection is less efficient and the ensuing crowding results in a lowering of lipid diffusion. These variations in lipid dynamics are corroborated by macroscopic rheological response measurements of PFT bound vesicles. Our study correlates PFT oligomeric state transitions, membrane remodeling, and mechanical property variations, providing unique insights into the pore forming mechanisms of cholesterol-dependent cytolysins.
Collapse
Affiliation(s)
| | - K Ganapathy Ayappa
- Center for BioSystems Science and Engineering Bengaluru, India; Department of Chemical Engineering Bengaluru, India.
| | - Jaydeep K Basu
- Department of Physics, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
31
|
Liposomes Prevent In Vitro Hemolysis Induced by Streptolysin O and Lysenin. MEMBRANES 2021; 11:membranes11050364. [PMID: 34069894 PMCID: PMC8157566 DOI: 10.3390/membranes11050364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022]
Abstract
The need for alternatives to antibiotics in the fight against infectious diseases has inspired scientists to focus on antivirulence factors instead of the microorganisms themselves. In this respect, prior work indicates that tiny, enclosed bilayer lipid membranes (liposomes) have the potential to compete with cellular targets for toxin binding, hence preventing their biological attack and aiding with their clearance. The effectiveness of liposomes as decoy targets depends on their availability in the host and how rapidly they are cleared from the circulation. Although liposome PEGylation may improve their circulation time, little is known about how such a modification influences their interactions with antivirulence factors. To fill this gap in knowledge, we investigated regular and long-circulating liposomes for their ability to prevent in vitro red blood cell hemolysis induced by two potent lytic toxins, lysenin and streptolysin O. Our explorations indicate that both regular and long-circulating liposomes are capable of similarly preventing lysis induced by streptolysin O. In contrast, PEGylation reduced the effectiveness against lysenin-induced hemolysis and altered binding dynamics. These results suggest that toxin removal by long-circulating liposomes is feasible, yet dependent on the particular virulence factor under scrutiny.
Collapse
|
32
|
What Can Mushroom Proteins Teach Us about Lipid Rafts? MEMBRANES 2021; 11:membranes11040264. [PMID: 33917311 PMCID: PMC8067419 DOI: 10.3390/membranes11040264] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/25/2022]
Abstract
The lipid raft hypothesis emerged as a need to explain the lateral organization and behavior of lipids in the environment of biological membranes. The idea, that lipids segregate in biological membranes to form liquid-disordered and liquid-ordered states, was faced with a challenge: to show that lipid-ordered domains, enriched in sphingomyelin and cholesterol, actually exist in vivo. A great deal of indirect evidence and the use of lipid-binding probes supported this idea, but there was a lack of tools to demonstrate the existence of such domains in living cells. A whole new toolbox had to be invented to biochemically characterize lipid rafts and to define how they are involved in several cellular functions. A potential solution came from basic biochemical experiments in the late 1970s, showing that some mushroom extracts exert hemolytic activities. These activities were later assigned to aegerolysin-based sphingomyelin/cholesterol-specific cytolytic protein complexes. Recently, six sphingomyelin/cholesterol binding proteins from different mushrooms have been identified and have provided some insight into the nature of sphingomyelin/cholesterol-rich domains in living vertebrate cells. In this review, we dissect the accumulated knowledge and introduce the mushroom lipid raft binding proteins as molecules of choice to study the dynamics and origins of these liquid-ordered domains in mammalian cells.
Collapse
|
33
|
Resnik N, Tratnjek L, Kreft ME, Kisovec M, Aden S, Bedina Zavec A, Anderluh G, Podobnik M, Veranič P. Cytotoxic Activity of LLO Y406A Is Targeted to the Plasma Membrane of Cancer Urothelial Cells. Int J Mol Sci 2021; 22:ijms22073305. [PMID: 33805017 PMCID: PMC8037347 DOI: 10.3390/ijms22073305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 01/01/2023] Open
Abstract
Identification of novel agents for bladder cancer treatment is highly desirable due to the high incidence of tumor recurrence and the risk of progression to muscle-invasive disease. The key feature of the cholesterol-dependent toxin listeriolysin O mutant (LLO Y406A) is its preferential activity at pH 5.7, which could be exploited either directly for selective targeting of cancer cells or the release of accumulated therapeutics from acidic endosomes. Therefore, our goal was to compare the cytotoxic effect of LLO Y406A on cancer cells (RT4) and normal urothelial cells (NPU), and to identify which cell membranes are the primary target of LLO Y406A by viability assays, life-cell imaging, fluorescence, and electron microscopy. LLO Y406A decreased viability, altered cell morphology, provoked membrane blebbing, and induced apoptosis in RT4 cells, while it did not affect NPU cells. LLO Y406A did not cause endosomal escape in RT4 cells, while the plasma membrane of RT4 cells was revealed as the primary target of LLO Y406A. It has been concluded that LLO Y406A has the ability to selectively eliminate cancer urothelial cells through pore-forming activity at the plasma membrane, without cytotoxic effects on normal urothelial cells. This promising selective activity merits further testing as an anti-cancer agent.
Collapse
Affiliation(s)
- Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (N.R.); (L.T.); (M.E.K.)
| | - Larisa Tratnjek
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (N.R.); (L.T.); (M.E.K.)
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (N.R.); (L.T.); (M.E.K.)
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (S.A.); (A.B.Z.); (G.A.); (M.P.)
| | - Saša Aden
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (S.A.); (A.B.Z.); (G.A.); (M.P.)
| | - Apolonija Bedina Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (S.A.); (A.B.Z.); (G.A.); (M.P.)
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (S.A.); (A.B.Z.); (G.A.); (M.P.)
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (S.A.); (A.B.Z.); (G.A.); (M.P.)
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (N.R.); (L.T.); (M.E.K.)
- Correspondence: ; Tel.: +386-1-543-7682
| |
Collapse
|
34
|
Jia C, Shi J, Han T, Yu ACH, Qin P. Plasma Membrane Blebbing Dynamics Involved in the Reversibly Perforated Cell by Ultrasound-Driven Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:733-750. [PMID: 33358511 DOI: 10.1016/j.ultrasmedbio.2020.11.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The perforation of plasma membrane by ultrasound-driven microbubbles (i.e., sonoporation) provides a temporary window for transporting macromolecules into the cytoplasm that is promising with respect to drug delivery and gene therapy. To improve the efficacy of delivery while ensuring biosafety, membrane resealing and cell recovery are required to help sonoporated cells defy membrane injury and regain their normal function. Blebs are found to accompany the recovery of sonoporated cells. However, the spatiotemporal characteristics of blebs and the underlying mechanisms remain unclear. With a customized platform for ultrasound exposure and 2-D/3-D live single-cell imaging, localized membrane perforation was induced with ultrasound-driven microbubbles, and the cellular responses were monitored using multiple fluorescent probes. The results indicated that localized blebs undergoing four phases (nucleation, expansion, pausing and retraction) on a time scale of tens of seconds to minutes were specifically involved in the reversibly sonoporated cells. The blebs spatially correlated with the membrane perforation site and temporally lagged (about tens of seconds to minutes) the resealing of perforated membrane. Their diameter (about several microns) and lifetime (about tens of seconds to minutes) positively correlated with the degree of sonoporation. Further studies revealed that intracellular calcium transients might be an upstream signal for triggering blebbing nucleation; exocytotic lysosomes not only contributed to resealing of the perforated membrane, but also to the increasing bleb volume during expansion; and actin components accumulation facilitated bleb retraction. These results provide new insight into the short-term strategies that the sonoporated cell employs to recover on membrane perforation and to remodel membrane structure and a biophysical foundation for sonoporation-based therapy.
Collapse
Affiliation(s)
- Caixia Jia
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianmin Shi
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Han
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| | - Peng Qin
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
35
|
Mageswaran SK, Yang WY, Chakrabarty Y, Oikonomou CM, Jensen GJ. A cryo-electron tomography workflow reveals protrusion-mediated shedding on injured plasma membrane. SCIENCE ADVANCES 2021; 7:7/13/eabc6345. [PMID: 33771860 PMCID: PMC7997517 DOI: 10.1126/sciadv.abc6345] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 02/09/2021] [Indexed: 06/12/2023]
Abstract
Cryo-electron tomography (cryo-ET) provides structural context to molecular mechanisms underlying biological processes. Although straightforward to implement for studying stable macromolecular complexes, using it to locate short-lived structures and events can be impractical. A combination of live-cell microscopy, correlative light and electron microscopy, and cryo-ET will alleviate this issue. We developed a workflow combining the three to study the ubiquitous and dynamic process of shedding in response to plasma membrane damage in HeLa cells. We found filopodia-like protrusions enriched at damage sites and acting as scaffolds for shedding, which involves F-actin dynamics, myosin-1a, and vacuolar protein sorting 4B (a component of the 'endosomal sorting complex required for transport' machinery). Overall, shedding is more complex than current models of vesiculation from flat membranes. Its similarities to constitutive shedding in enterocytes argue for a conserved mechanism. Our workflow can also be adapted to study other damage response pathways and dynamic cellular events.
Collapse
Affiliation(s)
- Shrawan Kumar Mageswaran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan.
| | - Yogaditya Chakrabarty
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Catherine M Oikonomou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Grant J Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84604, USA
| |
Collapse
|
36
|
Sonoda Y, Kano F, Murata M. Applications of cell resealing to reconstitute microRNA loading to extracellular vesicles. Sci Rep 2021; 11:2900. [PMID: 33536479 PMCID: PMC7859222 DOI: 10.1038/s41598-021-82452-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/20/2021] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are cargo carried by extracellular vesicles (EVs) and are associated with cell-cell interactions. The response to the cellular environment, such as disease states, genetic/metabolic changes, or differences in cell type, highly regulates cargo sorting to EVs. However, morphological features during EV formation and secretion involving miRNA loading are unknown. This study developed a new method of EV loading using cell resealing and reconstituted the elementary miRNA-loading processes. Morphology, secretory response, and cellular uptake ability of EVs obtained from intact and resealed HeLa cells were comparable. Exogenously added soluble factors were introduced into multivesicular endosomes (MVEs) and their subsequent secretion to the extracellular region occurred in resealed HeLa cells. In addition, miRNA transport to MVEs and miRNA encapsulation to EVs followed a distinct pathway regulated by RNA-binding proteins, such as Argonaute and Y-box binding protein 1, depending on miRNA types. Our cell-resealing system can analyze disease-specific EVs derived from disease model cells, where pathological cytosol is introduced into cells. Thus, EV formation in resealed cells can be used not only to create a reconstitution system to give mechanistic insight into EV encapsulation but also for applications such as loading various molecules into EVs and identifying disease-specific EV markers.
Collapse
Affiliation(s)
- Yuki Sonoda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Fumi Kano
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan.
| |
Collapse
|
37
|
Pore-forming proteins: From defense factors to endogenous executors of cell death. Chem Phys Lipids 2020; 234:105026. [PMID: 33309552 DOI: 10.1016/j.chemphyslip.2020.105026] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Pore-forming proteins (PFPs) and small antimicrobial peptides (AMPs) represent a large family of molecules with the common ability to punch holes in cell membranes to alter their permeability. They play a fundamental role as infectious bacteria's defensive tools against host's immune system and as executors of endogenous machineries of regulated cell death in eukaryotic cells. Despite being highly divergent in primary sequence and 3D structure, specific folds of pore-forming domains have been conserved. In fact, pore formation is considered an ancient mechanism that takes place through a general multistep process involving: membrane partitioning and insertion, oligomerization and pore formation. However, different PFPs and AMPs assemble and form pores following different mechanisms that could end up either in the formation of protein-lined or protein-lipid pores. In this review, we analyze the current findings in the mechanism of action of different PFPs and AMPs that support a wide role of membrane pore formation in nature. We also provide the newest insights into the development of state-of-art techniques that have facilitated the characterization of membrane pores. To understand the physiological role of these peptides/proteins or develop clinical applications, it is essential to uncover the molecular mechanism of how they perforate membranes.
Collapse
|
38
|
Molecular Dynamics Study of Lipid and Cholesterol Reorganization Due to Membrane Binding and Pore Formation by Listeriolysin O. J Membr Biol 2020; 253:535-550. [DOI: 10.1007/s00232-020-00148-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/10/2020] [Indexed: 12/20/2022]
|
39
|
Panevska A, Skočaj M, Modic Š, Razinger J, Sepčić K. Aegerolysins from the fungal genus Pleurotus - Bioinsecticidal proteins with multiple potential applications. J Invertebr Pathol 2020; 186:107474. [PMID: 32971130 DOI: 10.1016/j.jip.2020.107474] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
Abstract
The aegerolysin proteins ostreolysin A6, pleurotolysin A2 and erylysin A are produced by mushrooms of the genus Pleurotus. These aegerolysins can interact specifically with sphingolipid-enriched membranes. In particular, they strongly bind insect cells and to artificial lipid membranes that contain physiologically relevant concentrations of the main invertebrate-specific sphingolipid, ceramide phosphoethanolamine. Moreover, the aegerolysins permeabilise these membranes when combined with their protein partner pleurotolysin B, which contains a membrane-attack-complex/perforin domain. These aegerolysin/ pleurotolysin B complexes show strong and selective toxicity towards western corn rootworm larvae and adults and Colorado potato beetle larvae. Their insecticidal activities arise through aegerolysin binding to ceramide phosphoethanolamine in the insect midgut. This mode of membrane binding is different from those described for similar aegerolysin-based complexes of bacterial origin (e.g., Cry34Ab1/Cry35Ab1), or other Bacillus thuringiensis proteinaceous crystal toxins, which associate with protein receptors. The ability of Pleurotus aegerolysins to specifically interact with sphingolipid-enriched domains in mammalian cells can be further exploited to visualize lipid rafts in living cells, and to treat certain types of tumours and metabolic disorders. Finally, these proteins can strongly enhance fruiting initiation of P. ostreatus even when applied externally. In this review, we summarise the current knowledge of the potential biotechnological and biomedical applications of the Pleurotus aegerolysins, either alone or when complexed with pleurotolysin B, with special emphasis on their bioinsecticidal effects.
Collapse
Affiliation(s)
- Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| | - Špela Modic
- Plant Protection Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia.
| | - Jaka Razinger
- Plant Protection Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| |
Collapse
|
40
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
41
|
Induction of erythrocyte microvesicles by Escherichia Coli Alpha hemolysin. Biochem J 2020; 476:3455-3473. [PMID: 31661116 DOI: 10.1042/bcj20190546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/03/2019] [Accepted: 10/29/2019] [Indexed: 01/11/2023]
Abstract
Alpha hemolysin (HlyA) is the major virulence factor of uropathogenic Escherichia coli (UPEC) strains. Once in circulation, a low concentration of the toxin induces an increase in intracellular calcium that activates calpains - which proteolyse cytoskeleton proteins - and also favours the exposure of phosphatidylserine (PS) in the outer leaflet of erythrocyte membranes. All these events are considered part of eryptosis, as well as the delivery of microvesicles (MVs). Within this context, we studied the delivery of MVs by erythrocytes treated with sublytic concentrations of HlyA and demonstrated that HlyA-treated erythrocytes secrete MVs of diameter ∼200 nm containing HlyA and PS by a mechanism involving an increment of intracellular calcium concentration and purinergic receptor activation. Despite the presence of toxin in their membrane, HlyA-MVs are not hemolytically active and do not induce ATP release in untreated erythrocytes, thus suggesting that the delivery of HlyA-MVs might act as a protective mechanism on the part of erythrocytes that removes the toxin from the membrane to prevent the spread of infection. Although erythrocytes have been found to eliminate denatured hemoglobin and several membrane proteins by shedding MVs, the present work has revealed for the first time that an exogenous protein, such as a toxin, is eliminated by this process. This finding sheds light on the mechanism of action of the toxin and serves to further elucidate the consequences of UPEC infection in patients exhibiting HlyA-related diseases.
Collapse
|
42
|
Helfield B, Chen X, Watkins SC, Villanueva FS. Transendothelial Perforations and the Sphere of Influence of Single-Site Sonoporation. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1686-1697. [PMID: 32402675 PMCID: PMC7293920 DOI: 10.1016/j.ultrasmedbio.2020.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/08/2020] [Accepted: 02/27/2020] [Indexed: 05/28/2023]
Abstract
Acoustically driven gas bubble cavitation locally concentrates energy and can result in physical phenomena including sonoluminescence and erosion. In biomedicine, ultrasound-driven microbubbles transiently increase plasma membrane permeability (sonoporation) to promote drug/gene delivery. Despite its potential, little is known about cellular response in the aftermath of sonoporation. In the work described here, using a live-cell approach, we assessed the real-time interplay between transendothelial perforations (∼30-60 s) up to 650 µm2, calcium influx, breaching of the local cytoskeleton and sonoporation resealing upon F-actin recruitment to the perforation site (∼5-10 min). Through biophysical modeling, we established the critical role of membrane line tension in perforation resealing velocity (10-30 nm/s). Membrane budding/shedding post-sonoporation was observed on complete perforation closure, yet successful pore repair does not mark the end of sonoporation: protracted cell mobility from 8 µs of ultrasound is observed up to 4 h post-treatment. Taken holistically, we established the biophysical context of endothelial sonoporation repair with application in drug/gene delivery.
Collapse
Affiliation(s)
- Brandon Helfield
- Department of Physics, Concordia University, Montreal, Quebec, Canada; Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Simon C Watkins
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Heart and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
43
|
Varadarajan V, Desikan R, Ayappa KG. Assessing the extent of the structural and dynamic modulation of membrane lipids due to pore forming toxins: insights from molecular dynamics simulations. SOFT MATTER 2020; 16:4840-4857. [PMID: 32421131 DOI: 10.1039/d0sm00086h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Infections caused by many virulent bacterial strains are triggered by the release of pore forming toxins (PFTs), which form oligomeric transmembrane pore complexes on the target plasma membrane. The spatial extent of the perturbation to the surrounding lipids during pore formation is relatively unexplored. Using all-atom molecular dynamics simulations, we investigate the changes in the structure and dynamics of lipids in a 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) lipid bilayer in the presence of contrasting PFTs. Cytolysin A (ClyA), an α toxin with its inserted wedge shaped bundle of inserted α helices, induces significant asymmetry across the membrane leaflets in comparison with α hemolysin (AHL), a β toxin. Despite the differences in hydrophobic mismatch and uniquely different topologies of the two oligomers, perturbations to lipid order as reflected in the tilt angle and order parameters and membrane thinning are short ranged, lying within ∼2.5 nm from the periphery of either pore complex, and commensurate with distances typically associated with van der Waals forces. In contrast, the spatial extent of perturbations to the lipid dynamics extends outward to at least 4 nm for both proteins, and the continuous survival probabilities reveal the presence of a tightly bound shell of lipids in this region. Displacement probability distributions show long tails and the distinctly non-Gaussian features reflect the induced dynamic heterogeneity. A detailed profiling of the protein-lipid contacts with tyrosine, tryptophan, lysine and arginine residues shows increased non-polar contacts in the cytoplasmic leaflet for both PFTs, with a higher number of atomic contacts in the case of AHL in the extracellular leaflet due to the mushroom-like topology of the pore complex. The short ranged nature of the perturbations observed in this simple one component membrane suggests inherent plasticity of membrane lipids enabling the recovery of the structure and membrane fluidity even in the presence of these large oligomeric transmembrane protein assemblies. This observation has implications in membrane repair processes such as budding or vesicle fusion events used to mitigate PFT virulence, where the underlying lipid dynamics and fluidity in the vicinity of the pore complex are expected to play an important role.
Collapse
Affiliation(s)
- Vadhana Varadarajan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore-560012, India.
| | | | | |
Collapse
|
44
|
Temporary Membrane Permeabilization via the Pore-Forming Toxin Lysenin. Toxins (Basel) 2020; 12:toxins12050343. [PMID: 32456013 PMCID: PMC7290483 DOI: 10.3390/toxins12050343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
Pore-forming toxins are alluring tools for delivering biologically-active, impermeable cargoes to intracellular environments by introducing large conductance pathways into cell membranes. However, the lack of regulation often leads to the dissipation of electrical and chemical gradients, which might significantly affect the viability of cells under scrutiny. To mitigate these problems, we explored the use of lysenin channels to reversibly control the barrier function of natural and artificial lipid membrane systems by controlling the lysenin's transport properties. We employed artificial membranes and electrophysiology measurements in order to identify the influence of labels and media on the lysenin channel's conductance. Two cell culture models: Jurkat cells in suspension and adherent ATDC5 cells were utilized to demonstrate that lysenin channels may provide temporary cytosol access to membrane non-permeant propidium iodide and phalloidin. Permeability and cell viability were assessed by fluorescence spectroscopy and microscopy. Membrane resealing by chitosan or specific media addition proved to be an effective way of maintaining cellular viability. In addition, we loaded non-permeant dyes into liposomes via lysenin channels by controlling their conducting state with multivalent metal cations. The improved control over membrane permeability might prove fruitful for a large variety of biological or biomedical applications that require only temporary, non-destructive access to the inner environment enclosed by natural and artificial membranes.
Collapse
|
45
|
Peropadre A, Hazen MJ, Pérez Martín JM, Fernández Freire P. An acute exposure to perfluorooctanoic acid causes non-reversible plasma membrane injury in HeLa cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 260:114008. [PMID: 31995777 DOI: 10.1016/j.envpol.2020.114008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/17/2019] [Accepted: 01/16/2020] [Indexed: 06/10/2023]
Abstract
Health and environmental risks regarding perfluorooctanoic acid, a well-known perfluorinated compound, are still a subject of great concern. Ubiquitous exposure and disparity of results make it difficult to determine the underlying mechanism of action, especially at the cellular level. This study proposes an experimental design to assess the reversibility of adverse effects after a one-time exposure to the compound, in comparison with other more conventional timings. Complementary endpoints including total protein content, neutral red uptake and MTT reduction tests along with division rates and microscopic observations were evaluated in HeLa cells. In addition, PFOA quantification inside the cells was performed. The cellular effects exerted after 24 h exposure to perfluorooctanoic acid are non-reversible after a 48 h recovery period. In addition, we describe for the first time the induction of plasma membrane blebbing and the activation of membrane repair mechanisms after recovery from non-cytotoxic treatments with the compound. This experimental design has provided relevant information regarding the toxicity of this perfluorinated compound, relating all the adverse effects detected to its interaction with the plasma membrane.
Collapse
Affiliation(s)
- Ana Peropadre
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - Maria José Hazen
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - José Manuel Pérez Martín
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain
| | - Paloma Fernández Freire
- Department of Biology (Lab A-110), Faculty of Sciences, Universidad Autónoma de Madrid, C/Darwin 2, 28049, Madrid, Spain.
| |
Collapse
|
46
|
von Hoven G, Qin Q, Neukirch C, Husmann M, Hellmann N. Staphylococcus aureus α-toxin: small pore, large consequences. Biol Chem 2020; 400:1261-1276. [PMID: 30951494 DOI: 10.1515/hsz-2018-0472] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
The small β-pore-forming α-toxin, also termed α-hemolysin or Hla is considered to be an important virulence factor of Staphylococcus aureus. Perforation of the plasma membrane (PM) by Hla leads to uncontrolled flux of ions and water. Already a small number of toxin pores seems to be sufficient to induce complex cellular responses, many of which depend on the efflux of potassium. In this article, we discuss the implications of secondary membrane lesions, for example, by endogenous channels, for Hla-mediated toxicity, for calcium-influx and membrane repair. Activation of purinergic receptors has been proposed to be a major contributor to the lytic effects of various pore forming proteins, but new findings raise doubts that this holds true for Hla. However, the recently discovered cellular pore forming proteins gasdermin D and Mixed lineage kinase domain-like pseudokinase (MLKL) which perforate the PM from the cytosolic side might contribute to both calcium-influx-dependent damage and membrane repair. Activation of endogenous pore forming proteins by Hla above a threshold concentration could explain the apparent dependence of pore characteristics on toxin concentrations. If secondary membrane damage in the aftermath of Hla-attack contributes significantly to overall PM permeability, it might be an interesting target for new therapeutic approaches.
Collapse
Affiliation(s)
- Gisela von Hoven
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 67, 55131 Mainz, Germany
| | - Qianqian Qin
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 67, 55131 Mainz, Germany
| | - Claudia Neukirch
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 67, 55131 Mainz, Germany
| | - Matthias Husmann
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 67, 55131 Mainz, Germany
| | - Nadja Hellmann
- Institute for Pharmacy and Biochemistry, Johannes Gutenberg-University Mainz, Johann-Joachim Becher-Weg 30, 55128 Mainz, Germany
| |
Collapse
|
47
|
Kunishige R, Kano F, Murata M. The cell resealing technique for manipulating, visualizing, and elucidating molecular functions in living cells. Biochim Biophys Acta Gen Subj 2020; 1864:129329. [DOI: 10.1016/j.bbagen.2019.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
|
48
|
Wu N, Cernysiov V, Davidson D, Song H, Tang J, Luo S, Lu Y, Qian J, Gyurova IE, Waggoner SN, Trinh VQH, Cayrol R, Sugiura A, McBride HM, Daudelin JF, Labrecque N, Veillette A. Critical Role of Lipid Scramblase TMEM16F in Phosphatidylserine Exposure and Repair of Plasma Membrane after Pore Formation. Cell Rep 2020; 30:1129-1140.e5. [PMID: 31995754 PMCID: PMC7104872 DOI: 10.1016/j.celrep.2019.12.066] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/25/2019] [Accepted: 12/17/2019] [Indexed: 01/01/2023] Open
Abstract
Plasma membrane damage and cell death during processes such as necroptosis and apoptosis result from cues originating intracellularly. However, death caused by pore-forming agents, like bacterial toxins or complement, is due to direct external injury to the plasma membrane. To prevent death, the plasma membrane has an intrinsic repair ability. Here, we found that repair triggered by pore-forming agents involved TMEM16F, a calcium-activated lipid scramblase also mutated in Scott's syndrome. Upon pore formation and the subsequent influx of intracellular calcium, TMEM16F induced rapid "lipid scrambling" in the plasma membrane. This response was accompanied by membrane blebbing, extracellular vesicle release, preserved membrane integrity, and increased cell viability. TMEM16F-deficient mice exhibited compromised control of infection by Listeria monocytogenes associated with a greater sensitivity of neutrophils to the pore-forming Listeria toxin listeriolysin O (LLO). Thus, the lipid scramblase TMEM16F is critical for plasma membrane repair after injury by pore-forming agents.
Collapse
Affiliation(s)
- Ning Wu
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada; Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology (HUST), Wuhan, China.
| | - Vitalij Cernysiov
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada
| | - Hua Song
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jianlong Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Yan Lu
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada
| | - Ivayla E Gyurova
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stephen N Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Vincent Quoc-Huy Trinh
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, QC, Canada
| | - Romain Cayrol
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, QC, Canada
| | - Ayumu Sugiura
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Heidi M McBride
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | | | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada; Department of Medicine, University of Montréal, Montréal, QC H3C3J7, Canada; Department of Microbiology, Infectious Diseases and Immunology, University of Montréal, Montréal, QC H3C3J7, Canada
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W1R7, Canada; Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC H3G 1Y6, Canada.
| |
Collapse
|
49
|
Vötsch D, Willenborg M, Oelemann WM, Brogden G, Valentin-Weigand P. Membrane Binding, Cellular Cholesterol Content and Resealing Capacity Contribute to Epithelial Cell Damage Induced by Suilysin of Streptococcus suis. Pathogens 2019; 9:pathogens9010033. [PMID: 31905867 PMCID: PMC7168673 DOI: 10.3390/pathogens9010033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/31/2022] Open
Abstract
Streptococcus (S.) suis is a major cause of economic losses in the pig industry worldwide and is an emerging zoonotic pathogen. One important virulence-associated factor is suilysin (SLY), a toxin that belongs to the family of cholesterol-dependent pore-forming cytolysins (CDC). However, the precise role of SLY in host–pathogen interactions is still unclear. Here, we investigated the susceptibility of different respiratory epithelial cells to SLY, including immortalized cell lines (HEp-2 and NPTr cells), which are frequently used in in vitro studies on S. suis virulence mechanisms, as well as primary porcine respiratory cells, which represent the first line of barrier during S. suis infections. SLY-induced cell damage was determined by measuring the release of lactate dehydrogenase after infection with a virulent S. suis serotype 2 strain, its isogenic SLY-deficient mutant strain, or treatment with the recombinant protein. HEp-2 cells were most susceptible, whereas primary epithelial cells were hardly affected by the toxin. This prompted us to study possible explanations for these differences. We first investigated the binding capacity of SLY using flow cytometry analysis. Since binding and pore-formation of CDC is dependent on the membrane composition, we also determined the cellular cholesterol content of the different cell types using TLC and HPLC. Finally, we examined the ability of those cells to reseal SLY-induced pores using flow cytometry analysis. Our results indicated that the amount of membrane-bound SLY, the cholesterol content of the cells, as well as their resealing capacity all affect the susceptibility of the different cells regarding the effects of SLY. These findings underline the differences of in vitro pathogenicity models and may further help to dissect the biological role of SLY during S. suis infections.
Collapse
Affiliation(s)
- Désirée Vötsch
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
| | - Maren Willenborg
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
| | - Walter M.R. Oelemann
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
- Departamento de Imunologia, Instituto de Microbiologia Paulo Góes, Universidade Federal do Rio de Janeiro (UFRJ), 21941-901 Rio de Janeiro, Brazil
| | - Graham Brogden
- Department of Physiological Chemistry, University for Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Peter Valentin-Weigand
- Institute for Microbiology, University of Veterinary Medicine Hannover, 30173 Hannover, Germany; (D.V.); (M.W.)
- Correspondence: ; Tel.: +49-(0)511-856-7362
| |
Collapse
|
50
|
Boucher E, Goldin-Blais L, Basiren Q, Mandato CA. Actin dynamics and myosin contractility during plasma membrane repair and restoration: Does one ring really heal them all? CURRENT TOPICS IN MEMBRANES 2019; 84:17-41. [PMID: 31610862 DOI: 10.1016/bs.ctm.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In order to survive daily insults, cells have evolved various mechanisms that detect, stabilize and repair damages done to their plasma membrane and cytoskeletal structures. Damage to the PM endangers wounded cells by exposing them to uncontrolled exchanges with the extracellular milieu. The processes and molecular machinery enabling PM repair are therefore at the center of the bulk of the investigations into single-cell repair program. Wounds are repaired by dynamically remodeling the composition and shape of the injured area through exocytosis-mediated release of intracellular membrane components to the wounded area, endocytosis-mediated removal of the injured area, or the shedding of the injury. The wound healing program of Xenopus oocytes and early Drosophila embryos is by contrast, mostly characterized by the rapid formation of a large membrane patch over the wound that eventually fuse with the plasma membrane which restores plasma membrane continuity and lead to the shedding of patch material into the extracellular space. Formation and contraction of actomyosin ring restores normal plasma membrane composition and organizes cytoskeletal repairs. The extend of the contributions of the cytoskeleton to the wound healing program of somatic cells have comparatively received little attention. This review offers a survey of the current knowledge on how actin dynamics, myosin-based contraction and other cytoskeletal structures affects PM and cortical cytoskeleton repair of somatic cells.
Collapse
Affiliation(s)
- Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Laurence Goldin-Blais
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Quentin Basiren
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|