101
|
Hossain KR, Turkewitz DR, Holt SA, Herson L, Brown LJ, Cornell BA, Curmi PMG, Valenzuela SM. A conserved GXXXG motif in the transmembrane domain of CLIC proteins is essential for their cholesterol-dependant membrane interaction. Biochim Biophys Acta Gen Subj 2019; 1863:1243-1253. [PMID: 31075359 DOI: 10.1016/j.bbagen.2019.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sterols have been reported to modulate conformation and hence the function of several membrane proteins. One such group is the Chloride Intracellular Ion Channel (CLIC) family of proteins. The CLIC protein family consists of six evolutionarily conserved protein members in vertebrates. These proteins exist as both monomeric soluble proteins and as membrane bound proteins. To date, the structure of their membrane-bound form remains unknown. In addition to several studies indicating cellular redox environment and pH as facilitators of CLIC1 insertion into membranes, we have also demonstrated that the spontaneous membrane insertion of CLIC1 is regulated by membrane cholesterol. METHOD We have performed Langmuir-film, Impedance Spectroscopy and Molecular Docking Simulations to study the role of this GXXXG motif in CLIC1 interaction with cholesterol. RESULTS Unlike CLIC1-wild-type protein, the G18A and G22A mutants, that form part of the GXXXG motif, showed much slower initial kinetics and lower ion channel activity compared to the native protein. This difference can be attributed to the significantly reduced membrane interaction and insertion rate of the mutant proteins and/or slower formation of the final membrane configuration of the mutant proteins once in the membrane. CONCLUSION In this study, our findings uncover the identification of a GXXXG motif in CLIC1, which likely serves as the cholesterol-binding domain, that facilitates the protein's membrane interaction and insertion. Furthermore, we were able to postulate a model by which CLIC1 can autonomously insert into membranes to form functional ion channels. GENERAL SIGNIFICANCE Members of the CLIC family of proteins demonstrate unusual structural and dual functional properties - as ion channels and enzymes. Elucidating how the CLIC proteins' interact with membranes, thus allowing them to switch between their soluble and membrane form, will provide key information as to a mechanism of moonlighting activity and a novel regulatory role for cholesterol in such a process.
Collapse
Affiliation(s)
- Khondker Rufaka Hossain
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Daniel R Turkewitz
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Stephen A Holt
- Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Height, Australian Centre for Neutron Scattering, New South Wales 2234, Australia
| | - Leonie Herson
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Louise J Brown
- Department of Molecular Sciences, Macquarie University, New South Wales 2109, Australia
| | - Bruce A Cornell
- Surgical Diagnostics Pty Ltd., Roseville, Sydney 2069, Australia; Institute for Biomedical Materials and Devices, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Paul M G Curmi
- School of Physics, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Institute for Biomedical Materials and Devices, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| |
Collapse
|
102
|
Ehling-Schulz M, Lereclus D, Koehler TM. The Bacillus cereus Group: Bacillus Species with Pathogenic Potential. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0032-2018. [PMID: 31111815 PMCID: PMC6530592 DOI: 10.1128/microbiolspec.gpp3-0032-2018] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
The Bacillus cereus group includes several Bacillus species with closely related phylogeny. The most well-studied members of the group, B. anthracis, B. cereus, and B. thuringiensis, are known for their pathogenic potential. Here, we present the historical rationale for speciation and discuss shared and unique features of these bacteria. Aspects of cell morphology and physiology, and genome sequence similarity and gene synteny support close evolutionary relationships for these three species. For many strains, distinct differences in virulence factor synthesis provide facile means for species assignment. B. anthracis is the causative agent of anthrax. Some B. cereus strains are commonly recognized as food poisoning agents, but strains can also cause localized wound and eye infections as well as systemic disease. Certain B. thuringiensis strains are entomopathogens and have been commercialized for use as biopesticides, while some strains have been reported to cause infection in immunocompromised individuals. In this article we compare and contrast B. anthracis, B. cereus, and B. thuringiensis, including ecology, cell structure and development, virulence attributes, gene regulation and genetic exchange systems, and experimental models of disease.
Collapse
Affiliation(s)
- Monika Ehling-Schulz
- Institute of Microbiology, Department of Pathology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Didier Lereclus
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Theresa M Koehler
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center - Houston, Houston, TX 77030
| |
Collapse
|
103
|
Arcanobacterium haemolyticum Utilizes Both Phospholipase D and Arcanolysin To Mediate Its Uptake into Nonphagocytic Cells. Infect Immun 2019; 87:IAI.00832-18. [PMID: 30745329 DOI: 10.1128/iai.00832-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/04/2019] [Indexed: 01/14/2023] Open
Abstract
Arcanobacterium haemolyticum is an emerging human pathogen that causes pharyngitis and wound infections. A few studies have suggested that A. haemolyticum is able to induce its uptake into nonphagocytic epithelial cells, but the bacterial factors associated with host cell invasion and the host cell processes involved have yet to be studied. We investigated how two A. haemolyticum virulence factors, arcanolysin (ALN) and phospholipase D (PLD), affect the ability of the bacteria to adhere to and subsequently invade Detroit 562 pharyngeal epithelial cells. The sphingomyelinase activity of phospholipase D was necessary to increase bacterial adherence, while the absence of a functional arcanolysin had no effect on A. haemolyticum adherence but did lead to a decrease in A. haemolyticum invasion into Detroit 562 cells. Because of the known roles of cholesterol-dependent cytolysins in disrupting calcium gradients and inducing F-actin-mediated bacterial internalization, we sought to determine whether ALN and PLD played a similar role in the ability of A. haemolyticum to invade nonphagocytic cells. Elimination of extracellular calcium and inhibition of the Arp2/3 complex or F-actin polymerization also caused a decrease in the ability of A. haemolyticum to invade Detroit 562 cells. Overall, our findings suggest that A. haemolyticum utilizes phospholipase D primarily for adherence and utilizes arcanolysin primarily for invasion into Detroit 562 cells in a process dependent on extracellular calcium and F-actin polymerization. Our work marks the first insight into how the individual activities of arcanolysin and phospholipase D affect A. haemolyticum host-pathogen interactions using the biologically relevant Detroit 562 cell line.
Collapse
|
104
|
Sharma P, Sharma N, Mishra P, Joseph J, Mishra DK, Garg P, Roy S. Differential Expression of Antimicrobial Peptides in Streptococcus pneumoniae Keratitis and STAT3-Dependent Expression of LL-37 by Streptococcus pneumoniae in Human Corneal Epithelial Cells. Pathogens 2019; 8:pathogens8010031. [PMID: 30845777 PMCID: PMC6470555 DOI: 10.3390/pathogens8010031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pneumoniae is the leading cause of bacterial keratitis in the developing world with a growing trend of acquiring resistance against various antibiotics. In the current study, we determined the expression of different antimicrobial peptides (AMPs) in response to S. pneumoniae in patients, as well as in primary and immortalized human corneal epithelial cells. We further focused on LL-37 and determined its expression in human cornea infected with S. pneumoniae and studied the killing ability of LL-37 against S. pneumoniae. The expression of AMPs was determined by quantitative PCR and the phosphorylation of signaling proteins was evaluated by immunoblot analysis. LL-37 expression was also determined by immunofluorescence and Western blot method and the killing ability of LL-37 against S. pneumoniae was determined by colony-forming units. Differential expression of antimicrobial peptides was observed in patients with S. pneumoniae keratitis. Although S. pneumoniae induced expression of the AMPs in human corneal epithelial cells (HCEC), it did not induce AMP expression in U937, a human monocyte cell line. S. pneumoniae also caused activation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB)and mitogen activated protein kinase (MAPK) pathways in corneal epithelial cells. LL-37 was found to be effective against both laboratory and clinical strains of S. pneumoniae. LL-37 induction by S. pneumoniae in human corneal epithelial cells was mediated by signal transducer and activator of transcription 3 (STAT3) activation, and inhibition of STAT3 activation significantly reduced LL-37 expression. Our study determines an extensive profile of AMPs expressed in the human cornea during S. pneumoniae infection, and suggests the potential of LL-37 to be developed as an alternative therapeutic intervention to fight increasing antibiotic resistance among bacteria.
Collapse
Affiliation(s)
- Prerana Sharma
- Prof. Brien Holden Eye Research Center, LV Prasad Eye Institute, Hyderabad 500034, India.
- Department of Animal Sciences, University of Hyderabad, Hyderabad 500046, India.
| | - Natalia Sharma
- Prof. Brien Holden Eye Research Center, LV Prasad Eye Institute, Hyderabad 500034, India.
| | - Priyasha Mishra
- Prof. Brien Holden Eye Research Center, LV Prasad Eye Institute, Hyderabad 500034, India.
| | - Joveeta Joseph
- Jhaveri Microbiology Centre, LV Prasad Eye Institute, Hyderabad 500034, India.
| | - Dilip K Mishra
- Pathology Department, LV Prasad Eye Institute, Hyderabad 500034, India.
| | - Prashant Garg
- Tej Kohli Cornea Institute, Hyderabad 500034, India.
| | - Sanhita Roy
- Prof. Brien Holden Eye Research Center, LV Prasad Eye Institute, Hyderabad 500034, India.
- Tej Kohli Cornea Institute, Hyderabad 500034, India.
| |
Collapse
|
105
|
Putra I, Rabiee B, Anwar KN, Gidfar S, Shen X, Babalooee M, Ghassemi M, Afsharkhamseh N, Bakhsh S, Missiakas D, Nezamabadi A, Milani B, Eslani M, Djalilian AR. Staphylococcus aureus alpha-hemolysin impairs corneal epithelial wound healing and promotes intracellular bacterial invasion. Exp Eye Res 2019; 181:263-270. [PMID: 30822400 DOI: 10.1016/j.exer.2019.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
Colonization by Staphylococcus aureus (S. aureus) has been implicated in many infectious and wound healing disorders. This study was performed to characterize the pathogenic role of S. aureus alpha-hemolysin (alpha-toxin) in corneal epithelial wound healing and infectious keratitis in the setting of a corneal wound. The effect of wild-type and isogenic Hla mutant (α-hemolysin gene deleted) S. aureus bacteria and conditioned media on corneal epithelial wound healing was tested in vitro using a scratch assay and in vivo using a murine epithelial debridement model. The invasiveness of wild-type and Hla mutant S. aureus was evaluated in vitro in human corneal epithelial cells and in vivo in a murine model of infectious keratitis following total epithelial debridement. S. aureus and its conditioned media significantly delayed epithelial wound closure both in vitro (P < 0.05) and in vivo (P < 0.05). The effect of S. aureus on wound healing was significantly diminished with the Hla mutant strain (P < 0.05). Likewise, compared to the wild-type strain, the Hla mutant strain demonstrated significantly reduced ability to invade corneal epithelial cells in vitro (P < 0.05) and infect murine corneas following total epithelial debridement in vivo (P < 0.05). In conclusion, S. aureus alpha-hemolysin plays a major role in the pathologic modulation of corneal epithelial wound healing and the intracellular invasion of the bacteria. Limiting colonization by S. aureus and/or blocking alpha-hemolysin may provide a therapeutic approach for corneal wound healing and infectious disorders.
Collapse
Affiliation(s)
- Ilham Putra
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Behnam Rabiee
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Khandaker N Anwar
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Sanaz Gidfar
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Xiang Shen
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Mehrdad Babalooee
- Division of Infectious Diseases, Department of Medicine, University of Illinois at Chicago, 808 S. Wood St., Suite 888 (MC 735), Chicago, IL, 60612, United States
| | - Mahmood Ghassemi
- Division of Infectious Diseases, Department of Medicine, University of Illinois at Chicago, 808 S. Wood St., Suite 888 (MC 735), Chicago, IL, 60612, United States
| | - Neda Afsharkhamseh
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Saaquib Bakhsh
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Dominique Missiakas
- Department of Microbiology, University of Chicago, 920 East 58th St., CLSC 1117, Chicago, IL, 60637, United States
| | - Ali Nezamabadi
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Behrad Milani
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Medi Eslani
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States
| | - Ali R Djalilian
- Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1905 W. Taylor St., L-213, Chicago, IL, 60612, United States.
| |
Collapse
|
106
|
Penkauskas T, Preta G. Biological applications of tethered bilayer lipid membranes. Biochimie 2019; 157:131-141. [DOI: 10.1016/j.biochi.2018.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
|
107
|
Buchan KD, Foster SJ, Renshaw SA. Staphylococcus aureus: setting its sights on the human innate immune system. MICROBIOLOGY-SGM 2019; 165:367-385. [PMID: 30625113 DOI: 10.1099/mic.0.000759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Staphylococcus aureus has colonized humans for at least 10 000 years, and today inhabits roughly a third of the population. In addition, S. aureus is a major pathogen that is responsible for a significant disease burden, ranging in severity from mild skin and soft-tissue infections to life-threatening endocarditis and necrotizing pneumonia, with treatment often hampered by resistance to commonly available antibiotics. Underpinning its versatility as a pathogen is its ability to evade the innate immune system. S. aureus specifically targets innate immunity to establish and sustain infection, utilizing a large repertoire of virulence factors to do so. Using these factors, S. aureus can resist phagosomal killing, impair complement activity, disrupt cytokine signalling and target phagocytes directly using proteolytic enzymes and cytolytic toxins. Although most of these virulence factors are well characterized, their importance during infection is less clear, as many display species-specific activity against humans or against animal hosts, including cows, horses and chickens. Several staphylococcal virulence factors display species specificity for components of the human innate immune system, with as few as two amino acid changes reducing binding affinity by as much as 100-fold. This represents a major issue for studying their roles during infection, which cannot be examined without the use of humanized infection models. This review summarizes the major factors S. aureus uses to impair the innate immune system, and provides an in-depth look into the host specificity of S. aureus and how this problem is being approached.
Collapse
Affiliation(s)
- Kyle D Buchan
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Simon J Foster
- 2Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Stephen A Renshaw
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
108
|
Lam JGT, Song C, Seveau S. High-throughput Measurement of Plasma Membrane Resealing Efficiency in Mammalian Cells. J Vis Exp 2019. [PMID: 30663635 DOI: 10.3791/58351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
In their physiological environment, mammalian cells are often subjected to mechanical and biochemical stresses that result in plasma membrane damage. In response to these damages, complex molecular machineries rapidly reseal the plasma membrane to restore its barrier function and maintain cell survival. Despite 60 years of research in this field, we still lack a thorough understanding of the cell resealing machinery. With the goal of identifying cellular components that control plasma membrane resealing or drugs that can improve resealing, we have developed a fluorescence-based high-throughput assay that measures the plasma membrane resealing efficiency in mammalian cells cultured in microplates. As a model system for plasma membrane damage, cells are exposed to the bacterial pore-forming toxin listeriolysin O (LLO), which forms large 30-50 nm diameter proteinaceous pores in cholesterol-containing membranes. The use of a temperature-controlled multi-mode microplate reader allows for rapid and sensitive spectrofluorometric measurements in combination with brightfield and fluorescence microscopy imaging of living cells. Kinetic analysis of the fluorescence intensity emitted by a membrane impermeant nucleic acid-binding fluorochrome reflects the extent of membrane wounding and resealing at the cell population level, allowing for the calculation of the cell resealing efficiency. Fluorescence microscopy imaging allows for the enumeration of cells, which constitutively express a fluorescent chimera of the nuclear protein histone 2B, in each well of the microplate to account for potential variations in their number and allows for eventual identification of distinct cell populations. This high-throughput assay is a powerful tool expected to expand our understanding of membrane repair mechanisms via screening for host genes or exogenously added compounds that control plasma membrane resealing.
Collapse
Affiliation(s)
- Jonathan G T Lam
- Department of Microbial Infection and Immunity, The Ohio State University; Department of Microbiology, The Ohio State University; Infectious Diseases Institute, The Ohio State University
| | - Chi Song
- Division of Biostatistics, College of Public Health, The Ohio State University
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University; Department of Microbiology, The Ohio State University; Infectious Diseases Institute, The Ohio State University;
| |
Collapse
|
109
|
Panatala R, Barbato S, Kozai T, Luo J, Kapinos LE, Lim RYH. Nuclear Pore Membrane Proteins Self-Assemble into Nanopores. Biochemistry 2019; 58:484-488. [DOI: 10.1021/acs.biochem.8b01179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Radhakrishnan Panatala
- Biozentrum and Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Suncica Barbato
- Biozentrum and Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Toshiya Kozai
- Biozentrum and Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Jinghui Luo
- Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Larisa E. Kapinos
- Biozentrum and Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Roderick Y. H. Lim
- Biozentrum and Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
110
|
Pharmacological Targeting of Pore-Forming Toxins as Adjunctive Therapy for Invasive Bacterial Infection. Toxins (Basel) 2018; 10:toxins10120542. [PMID: 30562923 PMCID: PMC6316385 DOI: 10.3390/toxins10120542] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/23/2022] Open
Abstract
For many of the most important human bacterial infections, invasive disease severity is fueled by the cell damaging and pro-inflammatory effects of secreted pore-forming toxins (PFTs). Isogenic PFT-knockout mutants, e.g., Staphylococcus aureus lacking α-toxin or Streptococcus pneumoniae deficient in pneumolysin, show attenuation in animal infection models. This knowledge has inspired multi-model investigations of strategies to neutralize PFTs or counteract their toxicity as a novel pharmacological approach to ameliorate disease pathogenesis in clinical disease. Promising examples of small molecule, antibody or nanotherapeutic drug candidates that directly bind and neutralize PFTs, block their oligomerization or membrane receptor interactions, plug establishment membrane pores, or boost host cell resiliency to withstand PFT action have emerged. The present review highlights these new concepts, with a special focus on β-PFTs produced by leading invasive human Gram-positive bacterial pathogens. Such anti-virulence therapies could be applied as an adjunctive therapy to antibiotic-sensitive and -resistant strains alike, and further could be free of deleterious effects that deplete the normal microflora.
Collapse
|
111
|
Kulma M, Kacprzyk-Stokowiec A, Traczyk G, Kwiatkowska K, Dadlez M. Fine-tuning of the stability of β-strands by Y181 in perfringolysin O directs the prepore to pore transition. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:110-122. [PMID: 30463694 DOI: 10.1016/j.bbamem.2018.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 05/15/2018] [Accepted: 08/19/2018] [Indexed: 11/29/2022]
Abstract
Perfringolysin O (PFO) is a toxic protein that forms β-barrel transmembrane pores upon binding to cholesterol-containing membranes. The formation of lytic pores requires conformational changes in PFO that lead to the conversion of water-soluble monomers into membrane-bound oligomers. Although the general outline of stepwise pore formation has been established, the underlying mechanistic details await clarification. To extend our understanding of the molecular mechanisms that control the pore formation, we compared the hydrogen-deuterium exchange patterns of PFO with its derivatives bearing mutations in the D3 domain. In the case of two of these mutations F318A, Y181A, known from previous work to lead to a decreased lytic activity, global destabilization of all protein domains was observed in their water-soluble forms. This was accompanied by local changes in D3 β-sheet, including unexpected stabilization of functionally important β1 strand in Y181A. In case of the double mutation (F318A/Y181A) that completely abolished the lytic activity, several local changes were retained, but the global destabilization effects of single mutations were reverted and hydrogen-deuterium exchange (HDX) pattern returned to PFO level. Strong structural perturbations were not observed in case of remaining variants in which other residues of the hydrophobic core of D3 domain were substituted by alanine. Our results indicate the existence in PFO of a well-tuned H-bonding network that maintains the stability of the D3 β-strands at appropriate level at each transformation step. F318 and Y181 moieties participate in this network and their role extends beyond their direct intermolecular interaction during oligomerization that was identified previously.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5A Pawinskiego St., 02-106 Warsaw, Poland
| | - Aleksandra Kacprzyk-Stokowiec
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5A Pawinskiego St., 02-106 Warsaw, Poland
| | - Gabriela Traczyk
- Department of Cell Biology, The Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Department of Cell Biology, The Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Michał Dadlez
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 5A Pawinskiego St., 02-106 Warsaw, Poland; Institute of Genetics and Biotechnology, Department of Biology, Warsaw University, 1 Miecznikowa St., 02-185 Warsaw, Poland.
| |
Collapse
|
112
|
The Role of Pneumococcal Virulence Factors in Ocular Infectious Diseases. Interdiscip Perspect Infect Dis 2018; 2018:2525173. [PMID: 30538741 PMCID: PMC6257906 DOI: 10.1155/2018/2525173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pneumoniae is a gram-positive, facultatively anaerobic pathogen that can cause severe infections such as pneumonia, meningitis, septicemia, and middle ear infections. It is also one of the top pathogens contributing to bacterial keratitis and conjunctivitis. Though two pneumococcal vaccines exist for the prevention of nonocular diseases, they do little to fully prevent ocular infections. This pathogen has several virulence factors that wreak havoc on the conjunctiva, cornea, and intraocular system. Polysaccharide capsule aids in the evasion of host complement system. Pneumolysin (PLY) is a cholesterol-dependent cytolysin that acts as pore-forming toxin. Neuraminidases assist in adherence and colonization by exposing cell surface receptors to the pneumococcus. Zinc metalloproteinases contribute to evasion of the immune system and disease severity. The main purpose of this review is to consolidate the multiple studies that have been conducted on several pneumococcal virulence factors and the role each plays in conjunctivitis, keratitis, and endophthalmitis.
Collapse
|
113
|
Zhang W, Wang H, Wang B, Zhang Y, Hu Y, Ma B, Wang J. Replacing the 238th aspartic acid with an arginine impaired the oligomerization activity and inflammation-inducing property of pyolysin. Virulence 2018; 9:1112-1125. [PMID: 30067143 PMCID: PMC6086297 DOI: 10.1080/21505594.2018.1491256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Trueperella pyogenes (T. pyogenes) is an important opportunistic pathogen. Pyolysin (PLO) importantly contributes to the pathogenicity of T. pyogenes. However, the relationship between the structure and function and the virulence of PLO is not well documented. In the current study, recombinant PLO (rPLO) and three rPLO mutants were prepared. rPLO D238R, a mutant with the 238th aspartic acid replaced with an arginine, showed impairment in oligomerization activity on cholesterol-containing liposome and pore-forming activity on sheep red blood cell membrane. Further study employing the prepared mutants confirmed that the pore-forming activity of PLO is essential for inducing excessive inflammation responses in mice by upregulating the expression levels of IL-1β, TNF-α, and IL-6. By contrast, rPLO P499F, another mutant with impaired cell membrane binding capacity, elicited an inflammation response that was dependent on pathogen-associated molecular pattern (PAMP) activity, given that the mutant significantly upregulated the expression of IL-10 in macrophages and in mice, whereas rPLO did not. Results indicated that domain 1 of the PLO molecule plays an important role in maintaining pore-forming activity. Moreover, the PLO pore-forming activity and not PAMP activity is responsible for the inflammation-inducing effect of PLO. The results of this study provided new information for research field on the structure, function, and virulence of PLO. Abbreviations: T. pyogenes: Trueperella pyogenes; PLO: Pyolysin; rPLO: recombinant PLO; PAMP: pathogen-associated molecular pattern; CDCs: cholesterol-dependent cytolysins; PLY: pneumolysin; NLRP3: NLR family pyrin domain containing protein 3; PRRs: pattern recognition receptors; Asp: aspartic acid; TLR4: Toll-like receptor 4; Arg: arginine; Asn: asparagine; IPTG: Isopropyl-β-d-thiogalactoside; PBS: phosphate-buffered saline; sRBCs: sheep red blood cells; TEM: Transmission electron microscopy; RBCM: red blood cell membrane; SDS-PAGE: sodium dodecyl sulfate–polyacrylamide gel electrophoresis; NC membrane: nitrocellulose membrane; SDS-AGE: dodecyl sulfate agarose gel electrophoresis; MDBK cells: Madin–Darby bovine kidney cells; RPMI-1640 medium: Roswell Park Memorial Institute-1640 medium; FBS: fetal bovine serum; BMDMs: bone marrow-derived macrophages; TNF-α: tumor necrosis factor α; IL-1β: interleukin-1β; IFN-γ: interferon-γ; TGF-β: transforming growth factor-β; ELISA: enzyme-linked immunosorbent assay
Collapse
Affiliation(s)
- Wenlong Zhang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Haili Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Bing Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Yue Zhang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Yunhao Hu
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Bo Ma
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Junwei Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| |
Collapse
|
114
|
Vezočnik V, Hodnik V, Sitar S, Okur HI, Tušek-Žnidarič M, Lütgebaucks C, Sepčić K, Kogej K, Roke S, Žagar E, Maček P. Kinetically Stable Triglyceride-Based Nanodroplets and Their Interactions with Lipid-Specific Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:8983-8993. [PMID: 29983071 DOI: 10.1021/acs.langmuir.8b02180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Understanding of the interactions between proteins and natural and artificially prepared lipid membrane surfaces and embedded nonpolar cores is important in studies of physiological processes and their pathologies and is applicable to nanotechnologies. In particular, rapidly growing interest in cellular droplets defines the need for simplified biomimetic lipid model systems to overcome in vivo complexity and variability. We present a protocol for the preparation of kinetically stable nanoemulsions with nanodroplets composed of sphingomyelin (SM) and cholesterol (Chol), as amphiphilic surfactants, and trioleoylglycerol (TOG), at various molar ratios. To prepare stable SM/Chol-coated monodisperse lipid nanodroplets, we modified a reverse phase evaporation method and combined it with ultrasonication. Lipid composition, ζ-potential, gyration and hydrodynamic radius, shape, and temporal stability of the lipid nanodroplets were characterized and compared to extruded SM/Chol large unilamellar vesicles. Lipid nanodroplets and large unilamellar vesicles with theoretical SM/Chol/TOG molar ratios of 1/1/4.7 and 4/1/11.7 were further investigated for the orientational order of their interfacial water molecules using a second harmonic scattering technique, and for interactions with the SM-binding and Chol-binding pore-forming toxins equinatoxin II and perfringolysin O, respectively. The surface characteristics (ζ-potential, orientational order of interfacial water molecules) and binding of these proteins to the nanodroplet SM/Chol monolayers were similar to those for the SM/Chol bilayers of the large unilamellar vesicles and SM/Chol Langmuir monolayers, in terms of their surface structures. We propose that such SM/Chol/TOG nanoparticles with the required lipid compositions can serve as experimental models for monolayer membrane to provide a system that imitates the natural lipid droplets.
Collapse
Affiliation(s)
- Valerija Vezočnik
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Simona Sitar
- Department of Polymer Chemistry and Technology , National Institute of Chemistry , Hajdrihova 19 , Ljubljana 1000 , Slovenia
| | - Halil I Okur
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | | | - Cornelis Lütgebaucks
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| | - Ksenija Kogej
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology , University of Ljubljana , Večna pot 113 , Ljubljana 1000 , Slovenia
| | - Sylvie Roke
- Laboratory for Fundamental BioPhotonics, Institute of Bio-Engineering, and Institute of Material Science, School of Engineering, and Lausanne Centre for Ultrafast Science , École Polytechnique Fédérale de Lausanne , CH-1015 Lausanne , Switzerland
| | - Ema Žagar
- Department of Polymer Chemistry and Technology , National Institute of Chemistry , Hajdrihova 19 , Ljubljana 1000 , Slovenia
| | - Peter Maček
- Department of Biology, Biotechnical Faculty , University of Ljubljana , Jamnikarjeva 101 , Ljubljana 1000 , Slovenia
| |
Collapse
|
115
|
Arcanobacterium haemolyticum Phospholipase D Enzymatic Activity Promotes the Hemolytic Activity of the Cholesterol-Dependent Cytolysin Arcanolysin. Toxins (Basel) 2018; 10:toxins10060213. [PMID: 29882842 PMCID: PMC6024514 DOI: 10.3390/toxins10060213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/14/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Arcanolysin, produced by the human pathogen Arcanobacterium haemolyticum, is a cholesterol-dependent cytolysin. To mediate the pore-formation process, arcanolysin is secreted by A. haemolyticum and then must interact with cholesterol embedded within a host membrane. However, arcanolysin must compete with membrane components, such as the phospholipid sphingomyelin, to interact with cholesterol and form pores. Cholesterol forms transient hydrogen bonds with the extracellular portion of sphingomyelin, shielding cholesterol from extracellular factors, including arcanolysin. A. haemolyticum also produces a sphingomyelin-specific phospholipase D, which removes the choline head from sphingomyelin, leaving cyclic-ceramide phosphate and eliminating the potential for cholesterol sequestration. We hypothesized that the enzymatic activity of phospholipase D decreases sphingomyelin-mediated cholesterol sequestration and increases cholesterol accessibility for arcanolysin. Using purified arcanolysin and phospholipase D, we demonstrate that the enzymatic activity of phospholipase D is necessary to promote arcanolysin-mediated hemolysis in both time- and concentration-dependent manners. Phospholipase D promotion of arcanolysin-mediated cytotoxicity was confirmed in Detroit 562 epithelial cells. Furthermore, we determined that incubating phospholipase D with erythrocytes corresponds with an increase in the amount of arcanolysin bound to host membranes. This observation suggests that phospholipase D promotes arcanolysin-mediated cytotoxicity by increasing the ability of arcanolysin to bind to a host membrane.
Collapse
|
116
|
Rasha IM, Mohamed AA, Heba MA. Virulence and antimicrobial susceptibility profile of Listeria monocytogenes isolated from frozen vegetables available in the Egyptian market. ACTA ACUST UNITED AC 2018. [DOI: 10.5897/ajmr2018.8794] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
117
|
Rampersaud R, Lewis EL, LaRocca TJ, Ratner AJ. Environmental pH modulates inerolysin activity via post-binding blockade. Sci Rep 2018; 8:1542. [PMID: 29367601 PMCID: PMC5784117 DOI: 10.1038/s41598-018-19994-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/11/2018] [Indexed: 11/29/2022] Open
Abstract
The cholesterol dependent cytolysins (CDCs) are a family of pore-forming toxins produced by a wide range of bacteria. Some CDCs are important virulence factors for their cognate organisms, but their activity must be tightly regulated to ensure they operate at appropriate times and within the appropriate subcellular compartments. pH-dependent activity has been described for several CDCs, but the mechanism of such regulation has been studied in depth only for listeriolysin O (LLO), which senses environmental pH through a triad of acidic residues that mediate protein unfolding. Here we present data supporting a distinct mechanism for pH-dependence for inerolysin (INY), the CDC produced by Lactobacillus iners. Inerolysin (INY) has an acidic pH optimum with loss of activity at neutral pH. INY pH-dependence is characterized by reversible loss of pore formation with preservation of membrane binding. Fluorescent membrane probe assays indicated that INY insertion into host cell membranes, but not oligomerization, was defective at neutral pH. These data support the existence of a newly appreciated form of CDC pH-dependence functioning at a late stage of pore formation.
Collapse
Affiliation(s)
- Ryan Rampersaud
- College of Physicians & Surgeons, Columbia University, New York, NY, USA.,Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Emma L Lewis
- College of Physicians & Surgeons, Columbia University, New York, NY, USA.,Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, USA
| | - Timothy J LaRocca
- College of Physicians & Surgeons, Columbia University, New York, NY, USA.,Department of Basic & Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Adam J Ratner
- College of Physicians & Surgeons, Columbia University, New York, NY, USA. .,Department of Pediatrics, New York University School of Medicine, New York, NY, USA. .,Department of Microbiology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
118
|
Sarangi NK, Basu JK. Pathways for creation and annihilation of nanoscale biomembrane domains reveal alpha and beta-toxin nanopore formation processes. Phys Chem Chem Phys 2018; 20:29116-29130. [DOI: 10.1039/c8cp05729j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Raft-like functional domains with putative sizes of 20–200 nm and which are evolving dynamically are believed to be the most crucial regions in cellular membranes which determine cell signaling and various functions of cells.
Collapse
Affiliation(s)
| | - Jaydeep Kumar Basu
- Department of Physics
- Indian Institute of Science
- Bangalore – 560 012
- India
| |
Collapse
|
119
|
Nakamura S, Suzuki S, Saito H, Nishiyama KI. Cholesterol blocks spontaneous insertion of membrane proteins into liposomes of phosphatidylcholine. J Biochem 2017; 163:313-319. [DOI: 10.1093/jb/mvx083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/20/2017] [Indexed: 02/01/2023] Open
Affiliation(s)
- Shota Nakamura
- Cryobiofrontier Research Center, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, 020-8550 Iwate, Japan
| | - Sonomi Suzuki
- Cryobiofrontier Research Center, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, 020-8550 Iwate, Japan
| | - Hiroaki Saito
- RIKEN Quantitative Biology Center, Suita, 565-0874 Osaka, Japan
| | - Ken-ichi Nishiyama
- Cryobiofrontier Research Center, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, 020-8550 Iwate, Japan
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, 020-8550 Iwate, Japan
| |
Collapse
|
120
|
Griffin S, Preta G, Sheldon IM. Inhibiting mevalonate pathway enzymes increases stromal cell resilience to a cholesterol-dependent cytolysin. Sci Rep 2017; 7:17050. [PMID: 29213055 PMCID: PMC5719056 DOI: 10.1038/s41598-017-17138-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/22/2017] [Indexed: 01/28/2023] Open
Abstract
Animal health depends on the ability of immune cells to kill invading pathogens, and on the resilience of tissues to tolerate the presence of pathogens. Trueperella pyogenes causes tissue pathology in many mammals by secreting a cholesterol-dependent cytolysin, pyolysin (PLO), which targets stromal cells. Cellular cholesterol is derived from squalene, which is synthesized via the mevalonate pathway enzymes, including HMGCR, FDPS and FDFT1. The present study tested the hypothesis that inhibiting enzymes in the mevalonate pathway to reduce cellular cholesterol increases the resilience of stromal cells to PLO. We first verified that depleting cellular cholesterol with methyl-β-cyclodextrin increased the resilience of stromal cells to PLO. We then used siRNA to deplete mevalonate pathway enzyme gene expression, and used pharmaceutical inhibitors, atorvastatin, alendronate or zaragozic acid to inhibit the activity of HMGCR, FDPS and FDFT1, respectively. These approaches successfully reduced cellular cholesterol abundance, but mevalonate pathway enzymes did not affect cellular resilience equally. Inhibiting FDFT1 was most effective, with zaragozic acid reducing the impact of PLO on cell viability. The present study provides evidence that inhibiting FDFT1 increases stromal cell resilience to a cholesterol-dependent cytolysin.
Collapse
Affiliation(s)
- Sholeem Griffin
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, United Kingdom
| | - Giulio Preta
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, United Kingdom.,Institute of Biochemistry, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania
| | - Iain Martin Sheldon
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, United Kingdom.
| |
Collapse
|
121
|
Pore-forming toxins in Cnidaria. Semin Cell Dev Biol 2017; 72:133-141. [DOI: 10.1016/j.semcdb.2017.07.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 01/05/2023]
|
122
|
Vasta GR, Amzel LM, Bianchet MA, Cammarata M, Feng C, Saito K. F-Type Lectins: A Highly Diversified Family of Fucose-Binding Proteins with a Unique Sequence Motif and Structural Fold, Involved in Self/Non-Self-Recognition. Front Immunol 2017; 8:1648. [PMID: 29238345 PMCID: PMC5712786 DOI: 10.3389/fimmu.2017.01648] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/10/2017] [Indexed: 12/25/2022] Open
Abstract
The F-type lectin (FTL) family is one of the most recent to be identified and structurally characterized. Members of the FTL family are characterized by a fucose recognition domain [F-type lectin domain (FTLD)] that displays a novel jellyroll fold ("F-type" fold) and unique carbohydrate- and calcium-binding sequence motifs. This novel lectin family comprises widely distributed proteins exhibiting single, double, or greater multiples of the FTLD, either tandemly arrayed or combined with other structurally and functionally distinct domains, yielding lectin subunits of pleiotropic properties even within a single species. Furthermore, the extraordinary variability of FTL sequences (isoforms) that are expressed in a single individual has revealed genetic mechanisms of diversification in ligand recognition that are unique to FTLs. Functions of FTLs in self/non-self-recognition include innate immunity, fertilization, microbial adhesion, and pathogenesis, among others. In addition, although the F-type fold is distinctive for FTLs, a structure-based search revealed apparently unrelated proteins with minor sequence similarity to FTLs that displayed the FTLD fold. In general, the phylogenetic analysis of FTLD sequences from viruses to mammals reveals clades that are consistent with the currently accepted taxonomy of extant species. However, the surprisingly discontinuous distribution of FTLDs within each taxonomic category suggests not only an extensive structural/functional diversification of the FTLs along evolutionary lineages but also that this intriguing lectin family has been subject to frequent gene duplication, secondary loss, lateral transfer, and functional co-option.
Collapse
Affiliation(s)
- Gerardo R. Vasta
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - L. Mario Amzel
- Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mario A. Bianchet
- Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Matteo Cammarata
- Department of Earth and Marine Sciences, University of Palermo, Palermo, Italy
| | - Chiguang Feng
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Keiko Saito
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD, United States
| |
Collapse
|
123
|
Interaction of Cholesterol with Perfringolysin O: What Have We Learned from Functional Analysis? Toxins (Basel) 2017; 9:toxins9120381. [PMID: 29168745 PMCID: PMC5744101 DOI: 10.3390/toxins9120381] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) constitute a family of pore-forming toxins secreted by Gram-positive bacteria. These toxins form transmembrane pores by inserting a large β-barrel into cholesterol-containing membranes. Cholesterol is absolutely required for pore-formation. For most CDCs, binding to cholesterol triggers conformational changes that lead to oligomerization and end in pore-formation. Perfringolysin O (PFO), secreted by Clostridium perfringens, is the prototype for the CDCs. The molecular mechanisms by which cholesterol regulates the cytolytic activity of the CDCs are not fully understood. In particular, the location of the binding site for cholesterol has remained elusive. We have summarized here the current body of knowledge on the CDCs-cholesterol interaction, with focus on PFO. We have employed sterols in aqueous solution to identify structural elements in the cholesterol molecule that are critical for its interaction with PFO. In the absence of high-resolution structural information, site-directed mutagenesis data combined with binding studies performed with different sterols, and molecular modeling are beginning to shed light on this interaction.
Collapse
|
124
|
Hossain KR, Holt SA, Le Brun AP, Al Khamici H, Valenzuela SM. X-ray and Neutron Reflectivity Study Shows That CLIC1 Undergoes Cholesterol-Dependent Structural Reorganization in Lipid Monolayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:12497-12509. [PMID: 29016141 DOI: 10.1021/acs.langmuir.7b02872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
CLIC1 belongs to the ubiquitous family of chloride intracellular ion channel proteins that are evolutionarily conserved across species. The CLICs are unusual in that they exist mainly as soluble proteins but possess the intriguing property of spontaneous conversion from the soluble to an integral membrane-bound form. This conversion is regulated by the membrane lipid composition, especially by cholesterol, together with external factors such as oxidation and pH. However, the precise physiological mechanism regulating CLIC1 membrane insertion is currently unknown. In this study, X-ray and neutron reflectivity experiments were performed to study the interaction of CLIC1 with different phospholipid monolayers prepared using POPC, POPE, or POPS with and without cholesterol in order to better understand the regulatory role of cholesterol in CLIC1 membrane insertion. Our findings demonstrate for the first time two different structural orientations of CLIC1 within phospholipid monolayers, dependent upon the absence or presence of cholesterol. In phospholipid monolayers devoid of cholesterol, CLIC1 was unable to insert into the lipid acyl chain region. However, in the presence of cholesterol, CLIC1 showed significant insertion within the phospholipid acyl chains occupying an area per protein molecule of 6-7 nm2 with a total CLIC1 thickness ranging from ∼50 to 56 Å across the entire monolayer. Our data strongly suggests that cholesterol not only facilitates the initial docking or binding of CLIC1 to the membrane but also promotes deeper penetration of CLIC1 into the hydrophobic tails of the lipid monolayer.
Collapse
Affiliation(s)
- Khondker R Hossain
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Stephen A Holt
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | - Anton P Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, New South Wales 2234, Australia
| | | | | |
Collapse
|
125
|
Desikan R, Maiti PK, Ayappa KG. Assessing the Structure and Stability of Transmembrane Oligomeric Intermediates of an α-Helical Toxin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:11496-11510. [PMID: 28930630 DOI: 10.1021/acs.langmuir.7b02277] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Protein membrane interactions play an important role in our understanding of diverse phenomena ranging from membrane-assisted protein aggregation to oligomerization and folding. Pore-forming toxins (PFTs) are the primary vehicle for infection by several strains of bacteria. These proteins which are expressed in a water-soluble form (monomers) bind to the target membrane and conformationally transform (protomers) and self-assemble to form a multimer transmembrane pore complex through a process of oligomerization. On the basis of the structure of the transmembrane domains, PFTs are broadly classified into β or α toxins. In contrast to β-PFTs, the paucity of available crystal structures coupled with the amphipathic nature of the transmembrane domains has hindered our understanding of α-PFT pore formation. In this article, we use molecular dynamics (MD) simulations to examine the process of pore formation of the bacterial α-PFT, cytolysin A from Escherichia coli (ClyA) in lipid bilayer membranes. Using atomistic MD simulations ranging from 50 to 500 ns, we show that transmembrane oligomeric intermediates or "arcs" form stable proteolipidic complexes consisting of protein arcs with toroidal lipids lining the free edges. By creating initial conditions where the lipids are contained within the arcs, we study the dynamics of spontaneous lipid evacuation and toroidal edge formation. This process occurs on the time scale of tens of nanoseconds, suggesting that once protomers oligomerize, transmembrane arcs are rapidly stabilized to form functional water channels capable of leakage. Using umbrella sampling with a coarse-grained molecular model, we obtain the free energy of insertion of a single protomer into the membrane. A single inserted protomer has a stabilization free energy of -52.9 ± 1.2 kJ/mol and forms a stable transmembrane water channel capable of leakage. Our simulations reveal that arcs are stable and viable intermediates that can occur during the pore-formation pathway for ClyA.
Collapse
Affiliation(s)
- Rajat Desikan
- Department of Chemical Engineering, ‡Centre for Condensed Matter Theory, Department of Physics, and §Centre for Biosystems Science and Engineering, Indian Institute of Science , Bengaluru, India 560012
| | - Prabal K Maiti
- Department of Chemical Engineering, ‡Centre for Condensed Matter Theory, Department of Physics, and §Centre for Biosystems Science and Engineering, Indian Institute of Science , Bengaluru, India 560012
| | - K Ganapathy Ayappa
- Department of Chemical Engineering, ‡Centre for Condensed Matter Theory, Department of Physics, and §Centre for Biosystems Science and Engineering, Indian Institute of Science , Bengaluru, India 560012
| |
Collapse
|
126
|
Investigation of the inhibition effect and mechanism of myricetin to Suilysin by molecular modeling. Sci Rep 2017; 7:11748. [PMID: 28924148 PMCID: PMC5603505 DOI: 10.1038/s41598-017-12168-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/04/2017] [Indexed: 01/27/2023] Open
Abstract
In the present study, the inhibitory effect and mechanism of myricetin, a natural flavonoid compound, in relation to Suilysin (SLY) were investigated through molecular dynamics simulations, mutational analysis and fluorescence-quenching assays. Myricetin is a potential inhibitor that does not exhibit antimicrobial activity but has been shown to inhibit SLY cytotoxicity. Molecular dynamics simulations and mutational analysis revealed that myricetin binds directly to SLY in the gap between domains 2 and 3, an important region for oligomerization and pore formation. The results of principal component analysis (PCA) indicated that the binding of myricetin in this gap region restricts the conformational transition of SLY from a monomer to an oligomer, thereby counteracting the haemolytic activity of SLY. This mechanism was verified using a haemolysis assay. These results demonstrated that myricetin is a strong candidate as a novel therapeutic agent for the treatment of Streptococcus suis infections.
Collapse
|
127
|
Chen S, Soehnlen M, Downes FP, Walker ED. Insights from the draft genome into the pathogenicity of a clinical isolate of Elizabethkingia meningoseptica Em3. Stand Genomic Sci 2017; 12:56. [PMID: 28932346 PMCID: PMC5602931 DOI: 10.1186/s40793-017-0269-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/08/2017] [Indexed: 12/02/2022] Open
Abstract
Elizabethkingia meningoseptica is an emerging, healthcare-associated pathogen causing a high mortality rate in immunocompromised patients. We report the draft genome sequence of E. meningoseptica Em3, isolated from sputum from a patient with multiple underlying diseases. The genome has a length of 4,037,922 bp, a GC-content 36.4%, and 3673 predicted protein-coding sequences. Average nucleotide identity analysis (>95%) assigned the bacterium to the species E. meningoseptica. Genome analysis showed presence of the curli formation and assembly operon and a gene encoding hemagglutinins, indicating ability to form biofilm. In vitro biofilm assays demonstrated that E. meningoseptica Em3 formed more biofilm than E. anophelis Ag1 and E. miricola Emi3, both lacking the curli operon. A gene encoding thiol-activated cholesterol-dependent cytolysin in E. meningoseptica Em3 (potentially involved in lysing host immune cells) was also absent in E. anophelis Ag1 and E. miricola Emi3. Strain Em3 showed α-hemolysin activity on blood agar medium, congruent with presence of hemolysin and cytolysin genes. Furthermore, presence of heme uptake and utilization genes demonstrated adaptations for bloodstream infections. Strain Em3 contained 12 genes conferring resistance to β-lactams, including β-lactamases class A, class B, and metallo-β-lactamases. Results of comparative genomic analysis here provide insights into the evolution of E. meningoseptica Em3 as a pathogen.
Collapse
Affiliation(s)
- Shicheng Chen
- Department of Microbiology and Molecular Genetics, Michigan State University, 2215 Biomedical and Physical Sciences Building, 567 Wilson Road, East Lansing, MI 48824-4320 USA
| | - Marty Soehnlen
- Michigan Department of Health and Human Services, Bureau of Laboratories, Lansing, MI 48906 USA
| | - Frances P Downes
- Biomedical Laboratory Diagnostics Program, Michigan State University, East Lansing, MI 48824 USA
| | - Edward D Walker
- Department of Microbiology and Molecular Genetics, Michigan State University, 2215 Biomedical and Physical Sciences Building, 567 Wilson Road, East Lansing, MI 48824-4320 USA
| |
Collapse
|
128
|
Degradation of nuclear Ubc9 induced by listeriolysin O is dependent on K + efflux. Biochem Biophys Res Commun 2017; 493:1115-1121. [PMID: 28911869 DOI: 10.1016/j.bbrc.2017.09.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/10/2017] [Indexed: 01/11/2023]
Abstract
Listeriolysin O (LLO) is a pore-forming toxin produced by L. monocytogenes, and is belonged to a protein family of cholesterol-dependent cytolysins (CDCs). Previous studies have demonstrated that LLO triggers Ubc9 degradation and disrupts host SUMOylation to facilitate bacterial infection. However, the underlying mechanism of Ubc9 degradation is unclear. Here we show that LLO-induced down-regulation of Ubc9 is independent of Ubc9-SUMO interaction, however, it may involve phosphorylation signaling. Additionally, LLO exerts its effects primarily on nuclear Ubc9 and this process is mediated by K+ efflux. Interestingly, for intracellular CDCs such as pneumolysin and suilysin, blockage of K+ efflux enhances degradation of nuclear Ubc9, suggesting that extracellular and intracellular pathogens may exploit different mechanisms to modulate host SUMOylation system. Furthermore, up-regulation of SUMOylation by stable expression of SUMO-1 or SUMO-2 shows a delay in membrane perforation by LLO, indicating that SUMO modification of host proteins may act at the frontline for the defense response against LLO. Taken together, our study provides insights to the understanding of host-pathogen interactions.
Collapse
|
129
|
Yang L, Zhang Y, Wang H, Ma B, Xu L, Wang J, Zhang W. Identification of B-cell linear epitopes in domains 1-3 of pyolysin of Trueperella pyogenes using polyclonal antibodies. Vet Microbiol 2017; 210:24-31. [PMID: 29103692 DOI: 10.1016/j.vetmic.2017.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/23/2017] [Accepted: 08/26/2017] [Indexed: 11/17/2022]
Abstract
Trueperella pyogenes is an important opportunistic pathogen. Pyolysin (PLO) makes important contributions to the pathogenicity of T. pyogenes. However, the structure and function of PLO has not been well documented. In the current study, epitopes in domain 1-3 of PLO have been mapped using rabbit anti-recombinant PLO (rPLO) polyclonal antibodies, and then the results were re-checked by using mouse and chicken anti-rPLO polyclonal antibodies, respectively. The results indicated that the region of aa 281-393 in PLO could not elicit antibodies against linear epitopes. A total of six B cell linear epitopes have been found in domain 1 of PLO. Two of the six epitopes (EP1 and EP2) were used to immunize mice and chicken. Chicken anti-EP1 and anti-EP2 serum and mouse anti-EP2 serum could react with rPLO and corresponding epitope polypeptide in western blot assay; however, only mouse anti-EP2 serum shows weak anti-hemolysis effect in the rPLO and sheep red blood system. Our results provide some new information to the research field of PLO structure and function.
Collapse
Affiliation(s)
- Lingxiao Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China
| | - Yue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China
| | - Haili Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China
| | - Bo Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China
| | - Li Xu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Junwei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China.
| | - Wenlong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China; Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China.
| |
Collapse
|
130
|
Abdelmaksoud AA, Girerd PH, Garcia EM, Brooks JP, Leftwich LM, Sheth NU, Bradley SP, Serrano MG, Fettweis JM, Huang B, Strauss JF, Buck GA, Jefferson KK. Association between statin use, the vaginal microbiome, and Gardnerella vaginalis vaginolysin-mediated cytotoxicity. PLoS One 2017; 12:e0183765. [PMID: 28846702 PMCID: PMC5573284 DOI: 10.1371/journal.pone.0183765] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/10/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Bacterial vaginosis (BV) is the leading dysbiosis of the vaginal microbiome. The pathways leading towards the development of BV are not well understood. Gardnerella vaginalis is frequently associated with BV. G. vaginalis produces the cholesterol-dependent cytolysin (CDC), vaginolysin, which can lyse a variety of human cells and is thought to play a role in pathogenesis. Because membrane cholesterol is required for vaginolysin to function, and because HMG-CoA reductase inhibitors (statins) affect not only serum levels of cholesterol but membrane levels as well, we hypothesized that statins might affect the vaginal microbiome. METHODS To investigate the relationship between use of the statins and the vaginal microbiome, we analyzed 16S rRNA gene taxonomic surveys performed on vaginal samples from 133 women who participated in the Vaginal Human Microbiome Project and who were taking statins at the time of sampling, 152 women who reported high cholesterol levels but were not taking statins, and 316 women who did not report high cholesterol. To examine the effect of statins on the cytolytic effect of vaginolysin, the cholesterol-dependent cytolysin (CDC) produced by Gardnerella vaginalis, we assessed the effect of simvastatin pretreatment of VK2E6/E7 vaginal epithelial cells on vaginolysin-mediated cytotoxicity. RESULTS The mean proportion of G. vaginalis among women taking statins was significantly lower relative to women not using statins. Women using statins had higher mean proportions of Lactobacillus crispatus relative to women with normal cholesterol levels, and higher levels of Lactobacillus jensenii relative to women with high cholesterol but not taking statins. In vitro, vaginal epithelial cells pretreated with simvastatin were relatively resistant to vaginolysin and this effect was inhibited by cholesterol. CONCLUSIONS In this cross-sectional study, statin use was associated with reduced proportions of G. vaginalis and greater proportions of beneficial lactobacilli within the vaginal microbiome. The negative association between statin use and G. vaginalis may be related to inhibition of vaginolysin function.
Collapse
Affiliation(s)
- Abdallah A. Abdelmaksoud
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Philippe H. Girerd
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Erin M. Garcia
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - J. Paul Brooks
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Lauren M. Leftwich
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Nihar U. Sheth
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Steven P. Bradley
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Myrna G. Serrano
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jennifer M. Fettweis
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Bernice Huang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Jerome F. Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gregory A. Buck
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kimberly K. Jefferson
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
131
|
Tankovic J, Timinskas A, Janulaitiene M, Zilnyte M, Baudel JL, Maury E, Zvirbliene A, Pleckaityte M. Gardnerella vaginalis bacteremia associated with severe acute encephalopathy in a young female patient. Anaerobe 2017; 47:132-134. [PMID: 28546029 DOI: 10.1016/j.anaerobe.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/18/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
Gardnerella vaginalis is a facultative anaerobic bacterium that inhabits the genitourinary tract of both healthy women and those with bacterial vaginosis. We report a case of G. vaginalis bacteremia associated with severe toxic encephalopathy in a young woman. Anaerobic blood cultures yielded pure growth of small gram-variable rods later identified as G. vaginalis by both rapid biochemical tests and 16S rRNA gene sequencing. The patient recovered after treatment with amoxicillin-clavulanate according to the in vitro susceptibility testing. The complete genome of G. vaginalis isolate from blood cultures was determined. In vitro G. vaginalis isolate produced elevated amounts of a pore-forming toxin vaginolysin compared to control G. vaginalis isolates. We hypothesize that this toxin, if produced in high amounts in blood, is able to disrupt the blood-brain barrier and exert a toxic activity on brain cells.
Collapse
Affiliation(s)
- Jacques Tankovic
- Service de Bactériologie, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France; Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France.
| | - Albertas Timinskas
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Migle Janulaitiene
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Milda Zilnyte
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Jean-Luc Baudel
- Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France; Service de Réanimation Médicale, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France.
| | - Eric Maury
- Université Pierre et Marie Curie, 4 Place Jussieu, Paris, France; Service de Réanimation Médicale, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, 184 rue du Faubourg Saint-Antoine, Paris, France.
| | - Aurelija Zvirbliene
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| | - Milda Pleckaityte
- Institute of Biotechnology, Vilnius University, Sauletekio 7, Vilnius LT-10257, Lithuania.
| |
Collapse
|
132
|
Grayson KM, Blevins LK, Oliver MB, Ornelles DA, Swords WE, Alexander-Miller MA. Activation-dependent modulation of Streptococcus pneumoniae-mediated death in human lymphocytes. Pathog Dis 2017; 75:2966467. [PMID: 28158464 DOI: 10.1093/femspd/ftx008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/30/2017] [Indexed: 01/27/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a leading cause of community-acquired pneumonia, with infants and the elderly exhibiting significant susceptibility to the development of severe disease. A growing body of evidence supports the ability of Spn to negatively regulate the host response to infection, e.g. the capacity to induce death in numerous cell types. However, our understanding of the ability of Spn to directly impact lymphocytes remains limited. In this study, we tested the hypothesis that lymphocyte type and activation state influences the susceptibility to pneumococcus-mediated death. We show that in the resting state, CD4+ T cells exhibit a modestly increased susceptibility to Spn-induced death compared to CD8+ T cells or NK cells. In the presence of activating stimuli, the situation most reflective of what would occur in vivo during infection, all subsets demonstrated a significant increase in sensitivity to Spn-mediated death. Importantly, the activated subsets diverged dramatically in susceptibility with natural killer cells exhibiting an 8.6-fold greater sensitivity to pneumococcal components compared to the T-cell subsets. These results significantly expand our understanding of the capacity for pneumococcus to negatively regulate lymphocytes.
Collapse
|
133
|
van Pee K, Neuhaus A, D'Imprima E, Mills DJ, Kühlbrandt W, Yildiz Ö. CryoEM structures of membrane pore and prepore complex reveal cytolytic mechanism of Pneumolysin. eLife 2017; 6. [PMID: 28323617 PMCID: PMC5437283 DOI: 10.7554/elife.23644] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/17/2017] [Indexed: 12/20/2022] Open
Abstract
Many pathogenic bacteria produce pore-forming toxins to attack and kill human cells. We have determined the 4.5 Å structure of the ~2.2 MDa pore complex of pneumolysin, the main virulence factor of Streptococcus pneumoniae, by cryoEM. The pneumolysin pore is a 400 Å ring of 42 membrane-inserted monomers. Domain 3 of the soluble toxin refolds into two ~85 Å β-hairpins that traverse the lipid bilayer and assemble into a 168-strand β-barrel. The pore complex is stabilized by salt bridges between β-hairpins of adjacent subunits and an internal α-barrel. The apolar outer barrel surface with large sidechains is immersed in the lipid bilayer, while the inner barrel surface is highly charged. Comparison of the cryoEM pore complex to the prepore structure obtained by electron cryo-tomography and the x-ray structure of the soluble form reveals the detailed mechanisms by which the toxin monomers insert into the lipid bilayer to perforate the target membrane.
Collapse
Affiliation(s)
- Katharina van Pee
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Alexander Neuhaus
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Edoardo D'Imprima
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Deryck J Mills
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Werner Kühlbrandt
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Özkan Yildiz
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| |
Collapse
|
134
|
Goleij Z, Mahmoodzadeh Hosseini H, Amin M, Halabian R, Imani Fooladi AA. Prokaryotic toxins provoke different types of cell deaths in the eukaryotic cells. TOXIN REV 2017. [DOI: 10.1080/15569543.2017.1294180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zoleikha Goleij
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| | | | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran and
| |
Collapse
|
135
|
Smith WS, Baker EJ, Holmes SE, Koster G, Hunt AN, Johnston DA, Flavell SU, Flavell DJ. Membrane cholesterol is essential for triterpenoid saponin augmentation of a saporin-based immunotoxin directed against CD19 on human lymphoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:993-1007. [PMID: 28235471 DOI: 10.1016/j.bbamem.2017.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 01/06/2023]
Abstract
Triterpenoid saponins from Saponinum Album (SA) exert potent lytic effects on eukaryotic cell plasma membranes and, when used at sub-lytic concentrations, significantly augment the cytotoxicity of saporin-based immunotoxins (IT). To help elucidate the mechanism(s) behind these two phenomena we investigated the role of cholesterol to both. Human Daudi lymphoma cells were lipid deprived using a combination of three different approaches. Following treatment, the total cellular lipid content was analyzed by electrospray ionization mass spectrometry (ESI-MS) and plasma membrane (PM) cholesterol content measured using the lipophilic fluorescent probe NR12S. Maximal lipid deprivation of cells resulted in a complete loss of sensitivity to lysis by SA. Similarly augmentation of the anti-CD19 immunotoxin (IT) BU12-SAPORIN by SA was lost but without a concomitant loss of intrinsic IT cytotoxicity. The lytic activity of SA was restored following incubation of lipid deprived Daudi cells with Synthecol or LDL. The augmentative effect of SA on IT cytotoxicity for Daudi cells was restored following repletion of PM cholesterol levels with LDL. NR12S fluorescence and ESI-MS analysis of cellular lipids demonstrated that restoration of SA lytic activity by Synthecol was entirely due to increased PM cholesterol levels. Restoration of cellular and PM cholesterol levels by LDL also restored the augmentative effect of SA for IT, an effect associated with repletion of PM cholesterol with minor changes in some phospholipid species. These results indicate that the lytic and IT augmentative properties of SA are cholesterol-dependent in contrast to intrinsic IT cytotoxicity that is at least partially cholesterol independent.
Collapse
Affiliation(s)
- Wendy S Smith
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - Ella J Baker
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom; Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Suzanne E Holmes
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - Grielof Koster
- NIHR Respiratory Biomedical Research Unit, UHS, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom; Clinical and Experimental Sciences, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Alan N Hunt
- Clinical and Experimental Sciences, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - David A Johnston
- Biomedical Imaging Unit, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Sopsamorn U Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom.
| |
Collapse
|
136
|
van Pee K, Mulvihill E, Müller DJ, Yildiz Ö. Unraveling the Pore-Forming Steps of Pneumolysin from Streptococcus pneumoniae. NANO LETTERS 2016; 16:7915-7924. [PMID: 27796097 DOI: 10.1021/acs.nanolett.6b04219] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Pneumolysin (PLY) is the main virulence factor of Streptococcus pneumoniae that causes pneumonia, meningitis, and invasive pneumococcal infection. PLY is produced as monomers, which bind to cholesterol-containing membranes, where they oligomerize into large pores. To investigate the pore-forming mechanism, we determined the crystal structure of PLY at 2.4 Å and used it to design mutants on the surface of monomers. Electron microscopy of liposomes incubated with PLY mutants revealed that several mutations interfered with ring formation. Mutants that formed incomplete rings or linear arrays had strongly reduced hemolytic activity. By high-resolution time-lapse atomic force microscopy of wild-type PLY, we observed two different ring-shaped complexes. Most of the complexes protruded ∼8 nm above the membrane surface, while a smaller number protruded ∼11 nm or more. The lower complexes were identified as pores or prepores by the presence or absence of a lipid bilayer in their center. The taller complexes were side-by-side assemblies of monomers of soluble PLY that represent an early form of the prepore. Our observations suggest a four-step mechanism of membrane attachment and pore formation by PLY, which is discussed in the context of recent structural models. The functional separation of these steps is necessary for the understanding how cholesterol-dependent cytolysins form pores and lyse cells.
Collapse
Affiliation(s)
- Katharina van Pee
- Department of Structural Biology, Max PIanck Institute of Biophysics , Max von Laue Strasse 3, 60438 Frankfurt am Main, Germany
| | - Estefania Mulvihill
- Department of Biosystems Science and Engineering, ETH Zurich , Mattenstrasse 26, 4058 Basel, Switzerland
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, ETH Zurich , Mattenstrasse 26, 4058 Basel, Switzerland
| | - Özkan Yildiz
- Department of Structural Biology, Max PIanck Institute of Biophysics , Max von Laue Strasse 3, 60438 Frankfurt am Main, Germany
| |
Collapse
|
137
|
Abstract
Bacterial pathogens utilize a multitude of methods to invade mammalian hosts, damage tissue sites, and thwart the immune system from responding. One essential component of these strategies for many bacterial pathogens is the secretion of proteins across phospholipid membranes. Secreted proteins can play many roles in promoting bacterial virulence, from enhancing attachment to eukaryotic cells, to scavenging resources in an environmental niche, to directly intoxicating target cells and disrupting their functions. Many pathogens use dedicated protein secretion systems to secrete virulence factors from the cytosol of the bacteria into host cells or the host environment. In general, bacterial protein secretion apparatuses can be divided into classes, based on their structures, functions, and specificity. Some systems are conserved in all classes of bacteria and secrete a broad array of substrates, while others are only found in a small number of bacterial species and/or are specific to only one or a few proteins. In this chapter, we review the canonical features of several common bacterial protein secretion systems, as well as their roles in promoting the virulence of bacterial pathogens. Additionally, we address recent findings that indicate that the innate immune system of the host can detect and respond to the presence of protein secretion systems during mammalian infection.
Collapse
|
138
|
Munsell EV, Ross NL, Sullivan MO. Journey to the Center of the Cell: Current Nanocarrier Design Strategies Targeting Biopharmaceuticals to the Cytoplasm and Nucleus. Curr Pharm Des 2016; 22:1227-44. [PMID: 26675220 DOI: 10.2174/1381612822666151216151420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/15/2015] [Indexed: 01/06/2023]
Abstract
New biopharmaceutical molecules, potentially able to provide more personalized and effective treatments, are being identified through the advent of advanced synthetic biology strategies, sophisticated chemical synthesis approaches, and new analytical methods to assess biological potency. However, translation of many of these structures has been significantly limited due to the need for more efficient strategies to deliver macromolecular therapeutics to desirable intracellular sites of action. Engineered nanocarriers that encapsulate peptides, proteins, or nucleic acids are generally internalized into target cells via one of several endocytic pathways. These nanostructures, entrapped within endosomes, must navigate the intracellular milieu to orchestrate delivery to the intended destination, typically the cytoplasm or nucleus. For therapeutics active in the cytoplasm, endosomal escape continues to represent a limiting step to effective treatment, since a majority of nanocarriers trapped within endosomes are ultimately marked for enzymatic degradation in lysosomes. Therapeutics active in the nucleus have the added challenges of reaching and penetrating the nuclear envelope, and nuclear delivery remains a preeminent challenge preventing clinical translation of gene therapy applications. Herein, we review cutting-edge peptide- and polymer-based design strategies with the potential to enable significant improvements in biopharmaceutical efficacy through improved intracellular targeting. These strategies often mimic the activities of pathogens, which have developed innate and highly effective mechanisms to penetrate plasma membranes and enter the nucleus of host cells. Understanding these mechanisms has enabled advances in synthetic peptide and polymer design that may ultimately improve intracellular trafficking and bioavailability, leading to increased access to new classes of biotherapeutics.
Collapse
Affiliation(s)
| | | | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, Delaware.
| |
Collapse
|
139
|
Al Khamici H, Hossain KR, Cornell BA, Valenzuela SM. Investigating Sterol and Redox Regulation of the Ion Channel Activity of CLIC1 Using Tethered Bilayer Membranes. MEMBRANES 2016; 6:membranes6040051. [PMID: 27941637 PMCID: PMC5192407 DOI: 10.3390/membranes6040051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 12/03/2022]
Abstract
The Chloride Intracellular Ion Channel (CLIC) family consists of six conserved proteins in humans. These are a group of enigmatic proteins, which adopt both a soluble and membrane bound form. CLIC1 was found to be a metamorphic protein, where under specific environmental triggers it adopts more than one stable reversible soluble structural conformation. CLIC1 was found to spontaneously insert into cell membranes and form chloride ion channels. However, factors that control the structural transition of CLIC1 from being an aqueous soluble protein into a membrane bound protein have yet to be adequately described. Using tethered bilayer lipid membranes and electrical impedance spectroscopy system, herein we demonstrate that CLIC1 ion channel activity is dependent on the type and concentration of sterols in bilayer membranes. These findings suggest that membrane sterols play an essential role in CLIC1’s acrobatic switching from a globular soluble form to an integral membrane form, promoting greater ion channel conductance in membranes. What remains unclear is the precise nature of this regulation involving membrane sterols and ultimately determining CLIC1’s membrane structure and function as an ion channel. Furthermore, our impedance spectroscopy results obtained using CLIC1 mutants, suggest that the residue Cys24 is not essential for CLIC1’s ion channel function. However Cys24 does appear important for optimal ion channel activity. We also observe differences in conductance between CLIC1 reduced and oxidized forms when added to our tethered membranes. Therefore, we conclude that both membrane sterols and redox play a role in the ion channel activity of CLIC1.
Collapse
Affiliation(s)
- Heba Al Khamici
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
| | - Khondher R Hossain
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation (ANSTO), NSW 2234, Australia.
| | | | - Stella M Valenzuela
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
| |
Collapse
|
140
|
Ullah I, Chung K, Beloor J, Kim J, Cho M, Kim N, Lee KY, Kumar P, Lee SK. Trileucine residues in a ligand-CPP-based siRNA delivery platform improve endosomal escape of siRNA. J Drug Target 2016; 25:320-329. [PMID: 27820977 DOI: 10.1080/1061186x.2016.1258566] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
siRNA entrapment within endosomes is a significant problem encountered with siRNA delivery platforms that co-opt receptor-mediated entry pathways. Attachment of a cell-penetrating peptide (CPP), such as nona-arginine (9R) to a cell receptor-binding ligand like the Rabies virus glycoprotein, RVG, allows effective siRNA delivery to the cytoplasm by non-endocytic pathways, but a significant amount of siRNA complexes also enters the cell by ligand-induced receptor endocytosis and remain localized in endosomes. Here, we report that the incorporation of trileucine (3 Leu) residues as an endo-osmolytic moiety in the peptide improves endosomal escape and intracellular delivery of siRNA. The trileucine motif did not affect early non-endosomal mechanism of cytoplasmic siRNA delivery but enhanced target gene silencing by >20% only beyond 24 h of transfection when siRNA delivery is mostly through the endocytic route and siRNA trapped in the endosomes at later stages were subject to release into cytoplasm. The mechanism may involve endosomal membrane disruption as trileucine residues lysed RBCs selectively under endosomal pH conditions. Interestingly <3 Leu or >3 Leu residues were not as effective, suggesting that 3 Leu residues are useful for enhancing cytoplasmic delivery of siRNA routed through endosomes.
Collapse
Affiliation(s)
- Irfan Ullah
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Kunho Chung
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea.,b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Jagadish Beloor
- b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Jongkil Kim
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Minyoung Cho
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Nahyun Kim
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Kuen Yong Lee
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Priti Kumar
- b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Sang-Kyung Lee
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| |
Collapse
|
141
|
Sarangi NK, P II, Ayappa KG, Visweswariah SS, Basu JK. Super-resolution Stimulated Emission Depletion-Fluorescence Correlation Spectroscopy Reveals Nanoscale Membrane Reorganization Induced by Pore-Forming Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:9649-57. [PMID: 27564541 DOI: 10.1021/acs.langmuir.6b01848] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Membrane-protein interactions play a central role in membrane mediated cellular processes ranging from signaling, budding, and fusion, to transport across the cell membrane. Of particular significance is the process of efficient protein olgomerization and transmembrane pore formation on the membrane surface; the primary virulent pathway for the action of antimicrobial peptides and pore forming toxins (PFTs). The suggested nanoscopic length scales and dynamic nature of such membrane lipid-protein interactions makes their detection extremely challenging. Using a combination of super-resolution stimulated emission depletion nanoscopy with fluorescence correlation spectroscopy (STED-FCS) we unravel the emergence of nanoscale lateral heterogeneity in supported bilayer membranes made up of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and cholesterol upon interaction with the PFT, listeriolysin O (LLO). A distinct length scale-dependent dynamical crossover (<200 nm) from a Brownian diffusive regime is observed at 33 and 50% cholesterol compositions, indicating the partitioning of lipids into domains with variable cholesterol content. At 25% cholesterol content, this dyamical crossover is observed only in bilayers incubated with LLO providing evidence for the existence of sub ∼100 nm dynamical lipid nanodomains bound to LLO pore assemblies. By introducing asymmetry in cholesterol composition across the bilayer leaflets we infer that this domain formation is driven largely due to active cholesterol sequestration and transient trapping of lipids to the membrane bound motifs present in the toxins, en route to LLO oligomerization and subsequent pore formation. Bilayers prepared with labeled lipids present in either the proximal or distal leaflet allow us to track the dynamical perturbation in a leaflet-dependent manner upon LLO incubation. From the differences in the extent and intensity of the dynamical crossover as observed with STED-FCS, these experiments reveal that the affinity for cholesterol in the membrane binding motifs of the LLO subdomains induce cholesterol and lipid reorganization to a greater extent in the distal (upper) leaflet when compared with the proximal (lower) leaflet. The observed length scale-dependent membrane reorganization that occurs due to invasion by LLO could be generalized to other cholesterol-dependent cytolysins and emphasizes the significant advantage of using super-resolution STED nanoscopy to unravel complex lipid-protein interactions in membrane and cellular biophysics.
Collapse
Affiliation(s)
- Nirod Kumar Sarangi
- Department of Physics, ‡Center for Biosystems Science and Engineering, ¶Department of Chemical Engineering, and §Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560 012, India
| | - Ilanila I P
- Department of Physics, ‡Center for Biosystems Science and Engineering, ¶Department of Chemical Engineering, and §Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560 012, India
| | - K G Ayappa
- Department of Physics, ‡Center for Biosystems Science and Engineering, ¶Department of Chemical Engineering, and §Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560 012, India
| | - Sandhya S Visweswariah
- Department of Physics, ‡Center for Biosystems Science and Engineering, ¶Department of Chemical Engineering, and §Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560 012, India
| | - Jaydeep Kumar Basu
- Department of Physics, ‡Center for Biosystems Science and Engineering, ¶Department of Chemical Engineering, and §Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560 012, India
| |
Collapse
|
142
|
Lawrence SL, Gorman MA, Feil SC, Mulhern TD, Kuiper MJ, Ratner AJ, Tweten RK, Morton CJ, Parker MW. Structural Basis for Receptor Recognition by the Human CD59-Responsive Cholesterol-Dependent Cytolysins. Structure 2016; 24:1488-98. [PMID: 27499440 PMCID: PMC5320943 DOI: 10.1016/j.str.2016.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 11/23/2022]
Abstract
Cholesterol-dependent cytolysins (CDCs) are a family of pore-forming toxins that punch holes in the outer membrane of eukaryotic cells. Cholesterol serves as the receptor, but a subclass of CDCs first binds to human CD59. Here we describe the crystal structures of vaginolysin and intermedilysin complexed to CD59. These studies, together with small-angle X-ray scattering, reveal that CD59 binds to each at different, though overlapping, sites, consistent with molecular dynamics simulations and binding studies. The CDC consensus undecapeptide motif, which for the CD59-responsive CDCs has a proline instead of a tryptophan in the motif, adopts a strikingly different conformation between the structures; our data suggest that the proline acts as a selectivity switch to ensure CD59-dependent CDCs bind their protein receptor first in preference to cholesterol. The structural data suggest a detailed model of how these water-soluble toxins assemble as prepores on the cell surface.
Collapse
Affiliation(s)
- Sara L Lawrence
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Michael A Gorman
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Susanne C Feil
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Terrence D Mulhern
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J Kuiper
- Victorian Life Sciences Computation Initiative, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Adam J Ratner
- Departments of Pediatrics and Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma, Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Craig J Morton
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
143
|
Shoji A, Ikeya K, Aoyagi M, Takatsuji R, Yanagida A, Shibusawa Y, Sugawara M. Monitoring of cholesterol oxidation in a lipid bilayer membrane using streptolysin O as a sensing and signal transduction element. J Pharm Biomed Anal 2016; 128:455-461. [DOI: 10.1016/j.jpba.2016.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 11/26/2022]
|
144
|
Wolfmeier H, Radecke J, Schoenauer R, Koeffel R, Babiychuk VS, Drücker P, Hathaway LJ, Mitchell TJ, Zuber B, Draeger A, Babiychuk EB. Active release of pneumolysin prepores and pores by mammalian cells undergoing a Streptococcus pneumoniae attack. Biochim Biophys Acta Gen Subj 2016; 1860:2498-2509. [PMID: 27481675 DOI: 10.1016/j.bbagen.2016.07.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 07/15/2016] [Accepted: 07/24/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Streptococcus pneumoniae is a potent human pathogen. Its pore-forming exotoxin pneumolysin is instrumental for breaching the host's epithelial barrier and for the incapacitation of the immune system. METHODS AND RESULTS Using a combination of life imaging and cryo-electron microscopy we show that pneumolysin, released by cultured bacteria, is capable of permeabilizing the plasmalemma of host cells. However, such permeabilization does not lead to cell lysis since pneumolysin is actively removed by the host cells. The process of pore elimination starts with the formation of pore-bearing plasmalemmal nanotubes and proceeds by the shedding of pores that are embedded in the membrane of released microvesicles. Pneumolysin prepores are likewise removed. The protein composition of the toxin-induced microvesicles, assessed by mass spectrometry, is suggestive of a Ca(2+)-triggered mechanism encompassing the proteins of the annexin family and members of the endosomal sorting complex required for transport (ESCRT) complex. CONCLUSIONS S. pneumoniae releases sufficient amounts of pneumolysin to perforate the plasmalemma of host cells, however, the immediate cell lysis, which is frequently reported as a result of treatment with purified and artificially concentrated toxin, appears to be an unlikely event in vivo since the toxin pores are efficiently eliminated by microvesicle shedding. Therefore the dysregulation of cellular homeostasis occurring as a result of transient pore formation/elimination should be held responsible for the damaging toxin action. GENERAL SIGNIFICANCE We have achieved a comprehensive view of a general plasma membrane repair mechanism after injury by a major bacterial toxin.
Collapse
Affiliation(s)
- Heidi Wolfmeier
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Julika Radecke
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3000 Bern 9, Switzerland
| | - Roman Schoenauer
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - René Koeffel
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Viktoria S Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Patrick Drücker
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, Postfach, 3001, Bern, Switzerland
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Benoît Zuber
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland.
| | - Annette Draeger
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Eduard B Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland.
| |
Collapse
|
145
|
Tenenbaum T, Asmat TM, Seitz M, Schroten H, Schwerk C. Biological activities of suilysin: role in Streptococcus suis pathogenesis. Future Microbiol 2016; 11:941-54. [PMID: 27357518 DOI: 10.2217/fmb-2016-0028] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Streptococcus suis is an important swine and zoonotic pathogen equipped with several virulence factors. The pore-forming toxins are the most abundant bacterial toxins and classified as critical virulence (associated) factors of several pathogens. The role of suilysin (SLY), a pore-forming cholesterol-dependent cytolysin of S. suis, as a true virulence factor is under debate. Most of the bacterial toxins have been reported to modulate the host immune system to facilitate invasion and subsequent replication of bacteria within respective host cells. SLY has been demonstrated to play an important role in the pathogenesis of S. suis infection and inflammatory response in vitro and in vivo. This review highlights the contributions of SLY to the pathogenicity of S. suis. It will address its role during the development of S. suis meningitis in pigs, as well as humans, and discuss SLY as a potential vaccine candidate.
Collapse
Affiliation(s)
- Tobias Tenenbaum
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| | - Tauseef M Asmat
- Center for Advanced Studies in Vaccinology and Biotechnology, Brewery Road, University of Balochistan, 87300 Quetta, Pakistan
| | - Maren Seitz
- Institute for Microbiology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15, Hannover D-30173, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim D-68167, Germany
| |
Collapse
|
146
|
Gay A, Rye D, Radhakrishnan A. Switch-like responses of two cholesterol sensors do not require protein oligomerization in membranes. Biophys J 2016; 108:1459-1469. [PMID: 25809258 DOI: 10.1016/j.bpj.2015.02.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/27/2015] [Accepted: 02/12/2015] [Indexed: 10/23/2022] Open
Abstract
Many cellular processes are sensitive to levels of cholesterol in specific membranes and show a strongly sigmoidal dependence on membrane composition. The sigmoidal responses of the cholesterol sensors involved in these processes could arise from several mechanisms, including positive cooperativity (protein effects) and limited cholesterol accessibility (membrane effects). Here, we describe a sigmoidal response that arises primarily from membrane effects due to sharp changes in the chemical activity of cholesterol. Our models for eukaryotic membrane-bound cholesterol sensors are soluble bacterial toxins that show an identical switch-like specificity for endoplasmic reticulum membrane cholesterol. We show that truncated versions of these toxins fail to form oligomers but still show sigmoidal binding to cholesterol-containing membranes. The nonlinear response emerges because interactions between bilayer lipids control cholesterol accessibility to toxins in a threshold-like fashion. Around these thresholds, the affinity of toxins for membrane cholesterol varies by >100-fold, generating highly cooperative lipid-dependent responses independently of protein-protein interactions. Such lipid-driven cooperativity may control the sensitivity of many cholesterol-dependent processes.
Collapse
Affiliation(s)
- Austin Gay
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daphne Rye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
147
|
Ni T, Harlos K, Gilbert R. Structure of astrotactin-2: a conserved vertebrate-specific and perforin-like membrane protein involved in neuronal development. Open Biol 2016; 6:rsob.160053. [PMID: 27249642 PMCID: PMC4892435 DOI: 10.1098/rsob.160053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/07/2016] [Indexed: 11/30/2022] Open
Abstract
The vertebrate-specific proteins astrotactin-1 and 2 (ASTN-1 and ASTN-2) are integral membrane perforin-like proteins known to play critical roles in neurodevelopment, while ASTN-2 has been linked to the planar cell polarity pathway in hair cells. Genetic variations associated with them are linked to a variety of neurodevelopmental disorders and other neurological pathologies, including an advanced onset of Alzheimer's disease. Here we present the structure of the majority endosomal region of ASTN-2, showing it to consist of a unique combination of polypeptide folds: a perforin-like domain, a minimal epidermal growth factor-like module, a unique form of fibronectin type III domain and an annexin-like domain. The perforin-like domain differs from that of other members of the membrane attack complex-perforin (MACPF) protein family in ways that suggest ASTN-2 does not form pores. Structural and biophysical data show that ASTN-2 (but not ASTN-1) binds inositol triphosphates, suggesting a mechanism for membrane recognition or secondary messenger regulation of its activity. The annexin-like domain is closest in fold to repeat three of human annexin V and similarly binds calcium, and yet shares no sequence homology with it. Overall, our structure provides the first atomic-resolution description of a MACPF protein involved in development, while highlighting distinctive features of ASTN-2 responsible for its activity.
Collapse
Affiliation(s)
- Tao Ni
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Karl Harlos
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Robert Gilbert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
148
|
Castro-Gomes T, Corrotte M, Tam C, Andrews NW. Plasma Membrane Repair Is Regulated Extracellularly by Proteases Released from Lysosomes. PLoS One 2016; 11:e0152583. [PMID: 27028538 PMCID: PMC4814109 DOI: 10.1371/journal.pone.0152583] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/16/2016] [Indexed: 12/28/2022] Open
Abstract
Eukaryotic cells rapidly repair wounds on their plasma membrane. Resealing is Ca2+-dependent, and involves exocytosis of lysosomes followed by massive endocytosis. Extracellular activity of the lysosomal enzyme acid sphingomyelinase was previously shown to promote endocytosis and wound removal. However, whether lysosomal proteases released during cell injury participate in resealing is unknown. Here we show that lysosomal proteases regulate plasma membrane repair. Extracellular proteolysis is detected shortly after cell wounding, and inhibition of this process blocks repair. Conversely, surface protein degradation facilitates plasma membrane resealing. The abundant lysosomal cysteine proteases cathepsin B and L, known to proteolytically remodel the extracellular matrix, are rapidly released upon cell injury and are required for efficient plasma membrane repair. In contrast, inhibition of aspartyl proteases or RNAi-mediated silencing of the lysosomal aspartyl protease cathepsin D enhances resealing, an effect associated with the accumulation of active acid sphingomyelinase on the cell surface. Thus, secreted lysosomal cysteine proteases may promote repair by facilitating membrane access of lysosomal acid sphingomyelinase, which promotes wound removal and is subsequently downregulated extracellularly by a process involving cathepsin D.
Collapse
Affiliation(s)
- Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Christina Tam
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Norma W. Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
- * E-mail:
| |
Collapse
|
149
|
Bokori-Brown M, Petrov PG, Khafaji MA, Mughal MK, Naylor CE, Shore AC, Gooding KM, Casanova F, Mitchell TJ, Titball RW, Winlove CP. Red Blood Cell Susceptibility to Pneumolysin: CORRELATION WITH MEMBRANE BIOCHEMICAL AND PHYSICAL PROPERTIES. J Biol Chem 2016; 291:10210-27. [PMID: 26984406 DOI: 10.1074/jbc.m115.691899] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Indexed: 12/20/2022] Open
Abstract
This study investigated the effect of the biochemical and biophysical properties of the plasma membrane as well as membrane morphology on the susceptibility of human red blood cells to the cholesterol-dependent cytolysin pneumolysin, a key virulence factor of Streptococcus pneumoniae, using single cell studies. We show a correlation between the physical properties of the membrane (bending rigidity and surface and dipole electrostatic potentials) and the susceptibility of red blood cells to pneumolysin-induced hemolysis. We demonstrate that biochemical modifications of the membrane induced by oxidative stress, lipid scrambling, and artificial cell aging modulate the cell response to the toxin. We provide evidence that the diversity of response to pneumolysin in diabetic red blood cells correlates with levels of glycated hemoglobin and that the mechanical properties of the red blood cell plasma membrane are altered in diabetes. Finally, we show that diabetic red blood cells are more resistant to pneumolysin and the related toxin perfringolysin O relative to healthy red blood cells. Taken together, these studies indicate that the diversity of cell response to pneumolysin within a population of human red blood cells is influenced by the biophysical and biochemical status of the plasma membrane and the chemical and/or oxidative stress pre-history of the cell.
Collapse
Affiliation(s)
- Monika Bokori-Brown
- From the College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter EX4 4QD, United Kingdom,
| | - Peter G Petrov
- the College of Engineering, Mathematics and Physical Sciences, School of Physics, University of Exeter, Exeter EX4 4QL, United Kingdom
| | - Mawya A Khafaji
- the College of Engineering, Mathematics and Physical Sciences, School of Physics, University of Exeter, Exeter EX4 4QL, United Kingdom
| | - Muhammad K Mughal
- the Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Claire E Naylor
- the Department of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, United Kingdom
| | - Angela C Shore
- the Department of Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter EX2 5AX, United Kingdom, the National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter National Health Service Foundation Trust, Exeter EX2 5DW, United Kingdom, and
| | - Kim M Gooding
- the Department of Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter EX2 5AX, United Kingdom, the National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter National Health Service Foundation Trust, Exeter EX2 5DW, United Kingdom, and
| | - Francesco Casanova
- the Department of Diabetes and Vascular Medicine, University of Exeter Medical School, Barrack Road, Exeter EX2 5AX, United Kingdom, the National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter National Health Service Foundation Trust, Exeter EX2 5DW, United Kingdom, and
| | - Tim J Mitchell
- the Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Richard W Titball
- From the College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter EX4 4QD, United Kingdom
| | - C Peter Winlove
- the College of Engineering, Mathematics and Physical Sciences, School of Physics, University of Exeter, Exeter EX4 4QL, United Kingdom
| |
Collapse
|
150
|
Maekawa M, Yang Y, Fairn GD. Perfringolysin O Theta Toxin as a Tool to Monitor the Distribution and Inhomogeneity of Cholesterol in Cellular Membranes. Toxins (Basel) 2016; 8:toxins8030067. [PMID: 27005662 PMCID: PMC4810212 DOI: 10.3390/toxins8030067] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 11/23/2022] Open
Abstract
Cholesterol is an essential structural component of cellular membranes in eukaryotes. Cholesterol in the exofacial leaflet of the plasma membrane is thought to form membrane nanodomains with sphingolipids and specific proteins. Additionally, cholesterol is found in the intracellular membranes of endosomes and has crucial functions in membrane trafficking. Furthermore, cellular cholesterol homeostasis and regulation of de novo synthesis rely on transport via both vesicular and non-vesicular pathways. Thus, the ability to visualize and detect intracellular cholesterol, especially in the plasma membrane, is critical to understanding the complex biology associated with cholesterol and the nanodomains. Perfringolysin O (PFO) theta toxin is one of the toxins secreted by the anaerobic bacteria Clostridium perfringens and this toxin forms pores in the plasma membrane that causes cell lysis. It is well understood that PFO recognizes and binds to cholesterol in the exofacial leaflets of the plasma membrane, and domain 4 of PFO (D4) is sufficient for the binding of cholesterol. Recent studies have taken advantage of this high-affinity cholesterol-binding domain to create a variety of cholesterol biosensors by using a non-toxic PFO or the D4 in isolation. This review highlights the characteristics and usefulness of, and the principal findings related to, these PFO-derived cholesterol biosensors.
Collapse
Affiliation(s)
- Masashi Maekawa
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, 6th Floor, Toronto, ON M5S 1T8, Canada.
| | - Yanbo Yang
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, 6th Floor, Toronto, ON M5S 1T8, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, 6th Floor, Toronto, ON M5S 1T8, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada.
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Institute for Biomedical Engineering and Science Technology (IBEST), Ryerson University and St. Michael's Hospital, Toronto, ON M5B 2K3, Canada.
| |
Collapse
|