1
|
Rivero-Hernández AL, Hervis YP, Valdés-Tresanco ME, Escalona-Rodríguez FA, Cancelliere R, Relova-Hernández E, Romero-Hernández G, Pérez-Rivera E, Torres-Palacios Y, Cartaya-Quintero P, Ros U, Porchetta A, Micheli L, Fernández LE, Laborde R, Álvarez C, Sagan S, Lanio ME, Pazos Santos IF. Decoupling immunomodulatory properties from lipid binding in the α-pore-forming toxin Sticholysin II. Int J Biol Macromol 2024; 280:136244. [PMID: 39368578 DOI: 10.1016/j.ijbiomac.2024.136244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Sticholysin II (StII), a pore-forming toxin from the marine anemone Stichodactyla helianthus, enhances an antigen-specific cytotoxic T lymphocyte (CTL) response when co-encapsulated in liposomes with a model antigen. This capacity does not depend exclusively on its pore-forming activity and is partially supported by its ability to activate Toll-like receptor 4 (TLR4) in dendritic cells, presumably by interacting with this receptor or by triggering signaling cascades upon binding to lipid membrane. In order to investigate whether the lipid binding capacity of StII is required for immunomodulation, we designed a mutant in which the aromatic amino acids from the interfacial binding site Trp110, Tyr111 and Trp114 were substituted by Ala. In the present work, we demonstrated that StII3A keeps the secondary structure composition and global folding of StII, while it loses its lipid binding and permeabilization abilities. Despite this, StII3A upregulates dendritic cells maturation markers, enhances an antigen-specific effector CD8+ T cells response and confers antitumor protection in a preventive scenario in C57BL/6 mice. Our results indicate that a mechanism independent of its lipid binding ability is involved in the immunomodulatory capacity of StII, pointing to StII3A as a promising candidate to improve the reliability of the Sts-based vaccine platform.
Collapse
Affiliation(s)
- Ada L Rivero-Hernández
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Yadira P Hervis
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; Sorbonne Université, École normale supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
| | - Mario E Valdés-Tresanco
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; Center for Molecular Simulations and Department of Biological Sciences, University of Calgary, Alberta T2N 1N4, Canada.
| | - Felipe A Escalona-Rodríguez
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Rocco Cancelliere
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | | | - Glenda Romero-Hernández
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Eric Pérez-Rivera
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - Yusniel Torres-Palacios
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Patricia Cartaya-Quintero
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - Uris Ros
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany.
| | - Alessandro Porchetta
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | - Laura Micheli
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | | | - Rady Laborde
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Carlos Álvarez
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Sandrine Sagan
- Sorbonne Université, École normale supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France.
| | - Maria Eliana Lanio
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Isabel F Pazos Santos
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| |
Collapse
|
2
|
Gupta LK, Molla J, Prabhu AA. Story of Pore-Forming Proteins from Deadly Disease-Causing Agents to Modern Applications with Evolutionary Significance. Mol Biotechnol 2024; 66:1327-1356. [PMID: 37294530 DOI: 10.1007/s12033-023-00776-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/21/2023] [Indexed: 06/10/2023]
Abstract
Animal venoms are a complex mixture of highly specialized toxic molecules. Among them, pore-forming proteins (PFPs) or toxins (PFTs) are one of the major disease-causing toxic elements. The ability of the PFPs in defense and toxicity through pore formation on the host cell surface makes them unique among the toxin proteins. These features made them attractive for academic and research purposes for years in the areas of microbiology as well as structural biology. All the PFPs share a common mechanism of action for the attack of host cells and pore formation in which the selected pore-forming motifs of the host cell membrane-bound protein molecules drive to the lipid bilayer of the cell membrane and eventually produces water-filled pores. But surprisingly their sequence similarity is very poor. Their existence can be seen both in a soluble state and also in transmembrane complexes in the cell membrane. PFPs are prevalent toxic factors that are predominately produced by all kingdoms of life such as virulence bacteria, nematodes, fungi, protozoan parasites, frogs, plants, and also from higher organisms. Nowadays, multiple approaches to applications of PFPs have been conducted by researchers both in basic as well as applied biological research. Although PFPs are very devastating for human health nowadays researchers have been successful in making these toxic proteins into therapeutics through the preparation of immunotoxins. We have discussed the structural, and functional mechanism of action, evolutionary significance through dendrogram, domain organization, and practical applications for various approaches. This review aims to emphasize the PFTs to summarize toxic proteins together for basic knowledge as well as to highlight the current challenges, and literature gap along with the perspective of promising biotechnological applications for their future research.
Collapse
Affiliation(s)
- Laxmi Kumari Gupta
- Bioprocess Development Laboratory, Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India
| | - Johiruddin Molla
- Ghatal Rabindra Satabarsiki Mahavidyalaya Ghatal, Paschim Medinipur, Ghatal, West Bengal, 721212, India
| | - Ashish A Prabhu
- Bioprocess Development Laboratory, Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
3
|
Zimnyakov DA, Alonova MV, Lavrukhin MS, Lyapina AM, Feodorova VA. Polarization- and Chaos-Game-Based Fingerprinting of Molecular Targets of Listeria Monocytogenes Vaccine and Fully Virulent Strains. Curr Issues Mol Biol 2023; 45:10056-10078. [PMID: 38132474 PMCID: PMC10742786 DOI: 10.3390/cimb45120628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Two approaches to the synthesis of 2D binary identifiers ("fingerprints") of DNA-associated symbol sequences are considered in this paper. One of these approaches is based on the simulation of polarization-dependent diffraction patterns formed by reading the modeled DNA-associated 2D phase-modulating structures with a coherent light beam. In this case, 2D binarized distributions of close-to-circular extreme polarization states are applied as fingerprints of analyzed nucleotide sequences. The second approach is based on the transformation of the DNA-associated chaos game representation (CGR) maps into finite-dimensional binary matrices. In both cases, the differences between the structures of the analyzed and reference symbol sequences are quantified by calculating the correlation coefficient of the synthesized binary matrices. A comparison of the approaches under consideration is carried out using symbol sequences corresponding to nucleotide sequences of the hly gene from the vaccine and wild-type strains of Listeria monocytogenes as the analyzed objects. These strains differ in terms of the number of substituted nucleotides in relation to the vaccine strain selected as a reference. The results of the performed analysis allow us to conclude that the identification of structural differences in the DNA-associated symbolic sequences is significantly more efficient when using the binary distributions of close-to-circular extreme polarization states. The approach given can be applicable for genetic differentiation immunized from vaccinated animals (DIVA).
Collapse
Affiliation(s)
- Dmitry A. Zimnyakov
- Physics Department, Yury Gagarin State Technical University of Saratov, 77 Polytechnicheskaya Str., 410054 Saratov, Russia;
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering Named after N.I. Vavilov, 335 Sokolovaya Str., 410005 Saratov, Russia; (M.S.L.); (A.M.L.); (V.A.F.)
| | - Marina V. Alonova
- Physics Department, Yury Gagarin State Technical University of Saratov, 77 Polytechnicheskaya Str., 410054 Saratov, Russia;
| | - Maxim S. Lavrukhin
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering Named after N.I. Vavilov, 335 Sokolovaya Str., 410005 Saratov, Russia; (M.S.L.); (A.M.L.); (V.A.F.)
| | - Anna M. Lyapina
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering Named after N.I. Vavilov, 335 Sokolovaya Str., 410005 Saratov, Russia; (M.S.L.); (A.M.L.); (V.A.F.)
| | - Valentina A. Feodorova
- Laboratory for Fundamental and Applied Research, Saratov State University of Genetics, Biotechnology and Engineering Named after N.I. Vavilov, 335 Sokolovaya Str., 410005 Saratov, Russia; (M.S.L.); (A.M.L.); (V.A.F.)
- Department for Microbiology and Biotechnology, Saratov State University of Genetics, Biotechnology and Engineering Named after N.I. Vavilov, 335 Sokolovaya Str., 410005 Saratov, Russia
| |
Collapse
|
4
|
Wiktorczyk-Kapischke N, Skowron K, Wałecka-Zacharska E. Genomic and pathogenicity islands of Listeria monocytogenes-overview of selected aspects. Front Mol Biosci 2023; 10:1161486. [PMID: 37388250 PMCID: PMC10300472 DOI: 10.3389/fmolb.2023.1161486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Listeria monocytogenes causes listeriosis, a disease characterized by a high mortality rate (up to 30%). Since the pathogen is highly tolerant to changing conditions (high and low temperature, wide pH range, low availability of nutrients), it is widespread in the environment, e.g., water, soil, or food. L. monocytogenes possess a number of genes that determine its high virulence potential, i.e., genes involved in the intracellular cycle (e.g., prfA, hly, plcA, plcB, inlA, inlB), response to stress conditions (e.g., sigB, gadA, caspD, clpB, lmo1138), biofilm formation (e.g., agr, luxS), or resistance to disinfectants (e.g., emrELm, bcrABC, mdrL). Some genes are organized into genomic and pathogenicity islands. The islands LIPI-1 and LIPI-3 contain genes related to the infectious life cycle and survival in the food processing environment, while LGI-1 and LGI-2 potentially ensure survival and durability in the production environment. Researchers constantly have been searching for new genes determining the virulence of L. monocytogenes. Understanding the virulence potential of L. monocytogenes is an important element of public health protection, as highly pathogenic strains may be associated with outbreaks and the severity of listeriosis. This review summarizes the selected aspects of L. monocytogenes genomic and pathogenicity islands, and the importance of whole genome sequencing for epidemiological purposes.
Collapse
Affiliation(s)
- Natalia Wiktorczyk-Kapischke
- Department of Microbiology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Krzysztof Skowron
- Department of Microbiology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Ewa Wałecka-Zacharska
- Department of Food Hygiene and Consumer Health, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| |
Collapse
|
5
|
Gold Glyconanoparticles Combined with 91–99 Peptide of the Bacterial Toxin, Listeriolysin O, Are Efficient Immunotherapies in Experimental Bladder Tumors. Cancers (Basel) 2022; 14:cancers14102413. [PMID: 35626016 PMCID: PMC9140107 DOI: 10.3390/cancers14102413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/29/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary We propose a novel type of immunotherapy for bladder cancer using gold nanoparticles bound to a peptide of a bacterial toxin with anti-tumor capacities as listeriolysin O called Listeria nanovaccines. Here, we present the pre-clinical experiments on a mice model of bladder cancer and blood cells of patients with bladder cancer using these Listeria nanovaccines that activate the immune system, block the tumor immunosuppression environment, and reduce the tumor size. The impact of Listeria nanovaccines on the field of immunotherapies for solid tumors can be extended to other solid tumors containing lymphocyte infiltration. Therefore, we propose Listeria nanovaccines as immunotherapy for tumors such as melanoma, urothelial bladder carcinoma, non-small cell lung carcinoma, and glioblastoma. Abstract This study presents proof of concept assays to validate gold nanoparticles loaded with the bacterial peptide 91–99 of the listeriolysin O toxin (GNP-LLO91–99 nanovaccines) as immunotherapy for bladder tumors. GNP-LLO91–99 nanovaccines showed adjuvant abilities as they induce maturation and activation of monocyte-derived dendritic cells (MoDCs) to functional antigen-presenting cells in healthy donors and patients with melanoma or bladder cancer (BC), promoting a Th1 cytokine pattern. GNP-LLO91–99 nanovaccines were also efficient dendritic cell inducers of immunogenic tumor death using different bladder and melanoma tumor cell lines. The establishment of a pre-clinical mice model of subcutaneous BC confirmed that a single dose of GNP-LLO91–99 nanovaccines reduced tumor burden 4.7-fold and stimulated systemic Th1-type immune responses. Proof of concept assays validated GNP-LLO91–99 nanovaccines as immunotherapy by comparison to anti-CTLA-4 or anti-PD-1 antibodies. In fact, GNP-LLO91–99 nanovaccines increased percentages of CD4+ and CD8+ T cells, B cells, and functional antigen-presenting DCs in tumor-infiltrated lymphocytes, while they reduced the levels of myeloid-derived suppressor cells (MDSC) and suppressor T cells (Treg). We conclude that GNP-LLO91–99 nanovaccines can work as monotherapies or combinatory immunotherapies with anti-CTLA-4 or anti-PD-1 antibodies for solid tumors with high T cell infiltration, such as bladder cancer or melanoma.
Collapse
|
6
|
Petrova P, Arsov A, Tsvetanova F, Parvanova-Mancheva T, Vasileva E, Tsigoriyna L, Petrov K. The Complex Role of Lactic Acid Bacteria in Food Detoxification. Nutrients 2022; 14:2038. [PMID: 35631179 PMCID: PMC9147554 DOI: 10.3390/nu14102038] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Toxic ingredients in food can lead to serious food-related diseases. Such compounds are bacterial toxins (Shiga-toxin, listeriolysin, Botulinum toxin), mycotoxins (aflatoxin, ochratoxin, zearalenone, fumonisin), pesticides of different classes (organochlorine, organophosphate, synthetic pyrethroids), heavy metals, and natural antinutrients such as phytates, oxalates, and cyanide-generating glycosides. The generally regarded safe (GRAS) status and long history of lactic acid bacteria (LAB) as essential ingredients of fermented foods and probiotics make them a major biological tool against a great variety of food-related toxins. This state-of-the-art review aims to summarize and discuss the data revealing the involvement of LAB in the detoxification of foods from hazardous agents of microbial and chemical nature. It is focused on the specific properties that allow LAB to counteract toxins and destroy them, as well as on the mechanisms of microbial antagonism toward toxigenic producers. Toxins of microbial origin are either adsorbed or degraded, toxic chemicals are hydrolyzed and then used as a carbon source, while heavy metals are bound and accumulated. Based on these comprehensive data, the prospects for developing new combinations of probiotic starters for food detoxification are considered.
Collapse
Affiliation(s)
- Penka Petrova
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Alexander Arsov
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Flora Tsvetanova
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Tsvetomila Parvanova-Mancheva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Evgenia Vasileva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Lidia Tsigoriyna
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Kaloyan Petrov
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| |
Collapse
|
7
|
Malla BA, Ramanjeneya S, Vergis J, Malik SS, Barbuddhe SB, Rawool DB. Comparison of recombinant and synthetic listeriolysin- O peptide- based indirect ELISA vis-à-vis cultural isolation for detection of listeriosis in caprine and ovine species. J Microbiol Methods 2021; 188:106278. [PMID: 34246691 DOI: 10.1016/j.mimet.2021.106278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
The present study evaluated the comparative serodiagnostic efficacy of recombinant listeriolysin-O (rLLO) and synthetic LLO- 2 peptide-based indirect ELISA vis-à-vis cultural isolation using samples (n = 1326; blood, sera, vaginal swabs, and rectal swabs) collected from caprines (n = 350) and ovines (n = 50) having reproductive and/or nervous system disorders and/or healthy animals. On screening the test sera by rLLO- based ELISA, the antibodies against LLO (ALLO) were observed in 17.71% of the caprines and 2% of the ovines, respectively, while synthetic LLO-2- based ELISA revealed ALLO in 6.86% of caprines and not in ovines. Moreover, the adsorption of positive test sera with streptolysin-O (SLO) resulted in a significant reduction (7.43%; p < 0.05) in the seropositivity with rLLO- based ELISA, whereas LLO-2- based ELISA revealed marginal reduction (4.29%; p > 0.05) in the seropositivity. Overall, the seropositivity with LLO-2 synthetic peptide revealed comparatively less cross-reactivity in comparison to rLLO. The cultural isolation yielded five pathogenic L. monocytogenes isolates and three non-pathogenic Listeria spp. from caprine samples; however, Listeria spp. could not be recovered from any of the ovine samples. Further, on comparing seropositivity with the isolation study results, it was found that two out of the five animals from which pathogenic L. monocytogenes isolated were also found seropositive in both the ELISAs even after adsorption with SLO. Interestingly, rLLO- based ELISA detected antibodies against unadsorbed caprine sera even in those samples from which non-pathogenic Listeria spp. were isolated, whereas antibodies were not detected in LLO-2 peptide-based ELISA. In conclusion, it could be inferred that the synthetic LLO-2 peptide serves as a non- cross-reactive, ideal diagnostic antigen in serodiagnosis of capro-ovine listeriosis.
Collapse
Affiliation(s)
- Bilal Ahmad Malla
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, India
| | - Sunitha Ramanjeneya
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, India
| | - Jess Vergis
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, India
| | - Satyaveer Singh Malik
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, India
| | | | - Deepak Bhiwa Rawool
- Division of Veterinary Public Health, ICAR- Indian Veterinary Research Institute, Izatnagar, Bareilly 243 122, India; ICAR- National Research Centre on Meat, Chengicherla, Hyderabad 500 092, India.
| |
Collapse
|
8
|
Del Valle A, Acosta-Rivero N, Laborde RJ, Cruz-Leal Y, Cabezas S, Luzardo MC, Alvarez C, Labrada M, Rodríguez A, Rodríguez GL, Raymond J, Nogueira CV, Grubaugh D, Fernández LE, Higgins D, Lanio ME. Sticholysin II shows similar immunostimulatory properties to LLO stimulating dendritic cells and MHC-I restricted T cell responses of heterologous antigen. Toxicon 2021; 200:38-47. [PMID: 34237340 DOI: 10.1016/j.toxicon.2021.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Induction of CD8+ T cell responses against tumor cells and intracellular pathogens is an important goal of modern vaccinology. One approach of translational interest is the use of liposomes encapsulating pore-forming proteins (PFPs), such as Listeriolysin O (LLO), which has shown efficacy at priming strong and sustained CD8+ T cell responses. Recently, we have demonstrated that Sticholysin II (StII), a PFP from the sea anemone Stichodactyla helianthus, co-encapsulated into liposomes with ovalbumin (OVA) was able to stimulate, antigen presenting cells, antigen-specific CD8+ T cells and anti-tumor activity in mice. In the present study, we aimed to compare StII and LLO in terms of their abilities to stimulate dendritic cells and to induce major histocompatibility complex (MHC) class I restricted T cell responses against OVA. Interestingly, StII exhibited similar abilities to LLO in vitro of inducing dendritic cells maturation, as measured by increased expression of CD40, CD80, CD86 and MHC-class II molecules, and of stimulating OVA cross-presentation to a CD8+ T cell line. Remarkably, using an ex vivo Enzyme-Linked ImmunoSpot Assay (ELISPOT) to monitor gamma interferon (INF-γ) producing effector memory CD8+ T cells, liposomal formulations containing either StII or LLO induced comparable frequencies of OVA-specific INF-γ producing CD8+ T cells in mice that were sustained in time. However, StII-containing liposomes stimulated antigen-specific memory CD8+ T cells with a higher potential to secrete IFN-γ than liposomes encapsulating LLO. This StII immunostimulatory property further supports its use for the rational design of T cell vaccines against cancers and intracellular pathogens. In summary, this study indicates that StII has immunostimulatory properties similar to LLO, despite being evolutionarily distant PFPs.
Collapse
Affiliation(s)
- A Del Valle
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - N Acosta-Rivero
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba.
| | - R J Laborde
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - Y Cruz-Leal
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - S Cabezas
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - M C Luzardo
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - C Alvarez
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba
| | - M Labrada
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - A Rodríguez
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - G L Rodríguez
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - J Raymond
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | | | - D Grubaugh
- Harvard Medical School, Harvard University, USA
| | - L E Fernández
- Center of Molecular Immunology (CIM), Playa, La Habana, Cuba
| | - D Higgins
- Harvard Medical School, Harvard University, USA
| | - M E Lanio
- Center for Protein Studies, Faculty of Biology, Havana University (UH) and Lab UH-CIM, Cuba.
| |
Collapse
|
9
|
Soria-Castro R, Alfaro-Doblado ÁR, Rodríguez-López G, Campillo-Navarro M, Meneses-Preza YG, Galán-Salinas A, Alvarez-Jimenez V, Yam-Puc JC, Munguía-Fuentes R, Domínguez-Flores A, Estrada-Parra S, Pérez-Tapia SM, Chávez-Blanco AD, Chacón-Salinas R. TLR2 Regulates Mast Cell IL-6 and IL-13 Production During Listeria monocytogenes Infection. Front Immunol 2021; 12:650779. [PMID: 34194428 PMCID: PMC8238461 DOI: 10.3389/fimmu.2021.650779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022] Open
Abstract
Listeria monocytogenes (L.m) is efficiently controlled by several cells of the innate immunity, including the Mast Cell (MC). MC is activated by L.m inducing its degranulation, cytokine production and microbicidal mechanisms. TLR2 is required for the optimal control of L.m infection by different cells of the immune system. However, little is known about the MC receptors involved in recognizing this bacterium and whether these interactions mediate MC activation. In this study, we analyzed whether TLR2 is involved in mediating different MC activation responses during L.m infection. We found that despite MC were infected with L.m, they were able to clear the bacterial load. In addition, MC degranulated and produced ROS, TNF-α, IL-1β, IL-6, IL-13 and MCP-1 in response to bacterial infection. Interestingly, L.m induced the activation of signaling proteins: ERK, p38 and NF-κB. When TLR2 was blocked, L.m endocytosis, bactericidal activity, ROS production and mast cell degranulation were not affected. Interestingly, only IL-6 and IL-13 production were affected when TLR2 was inhibited in response to L.m infection. Furthermore, p38 activation depended on TLR2, but not ERK or NF-κB activation. These results indicate that TLR2 mediates only some MC activation pathways during L.m infection, mainly those related to IL-6 and IL-13 production.
Collapse
Affiliation(s)
- Rodolfo Soria-Castro
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Ángel R. Alfaro-Doblado
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Gloria Rodríguez-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Marcia Campillo-Navarro
- Research Coordination, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Yatsiri G. Meneses-Preza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Adrian Galán-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Violeta Alvarez-Jimenez
- Unidad de Citometría de Flujo, Lab de Biología Molecular y Bioseguridad Nivel 3, Centro Médico Naval, Secretaría de Marina (SEMAR), Mexico City, Mexico
| | - Juan C. Yam-Puc
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Rosario Munguía-Fuentes
- Departamento de Ciencias Básicas, Unidad Profesional Interdisciplinaria en Ingeniería y Tecnologías Avanzadas, Instituto Politécnico Nacional (UPIITA-IPN), Mexico City, Mexico
| | - Adriana Domínguez-Flores
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Sonia M. Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
- Unidad de Desarrollo e Investigación en Bioprocesos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| | - Alma D. Chávez-Blanco
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), México City, Mexico
| | - Rommel Chacón-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City, Mexico
| |
Collapse
|
10
|
Antilisterial Potential of Lactic Acid Bacteria in Eliminating Listeria monocytogenes in Host and Ready-to-Eat Food Application. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Listeriosis is a severe food borne disease with a mortality rate of up to 30% caused by pathogenic Listeria monocytogenes via the production of several virulence factors including listeriolysin O (LLO), transcriptional activator (PrfA), actin (Act), internalin (Int), etc. It is a foodborne disease predominantly causing infections through consumption of contaminated food and is often associated with ready-to-eat food (RTE) and dairy products. Common medication for listeriosis such as antibiotics might cause an eagle effect and antibiotic resistance if it is overused. Therefore, exploration of the use of lactic acid bacteria (LAB) with probiotic characteristics and multiple antimicrobial properties is increasingly getting attention for their capability to treat listeriosis, vaccine development, and hurdle technologies. The antilisterial gene, a gene coding to produce antimicrobial peptide (AMP), one of the inhibitory substances found in LAB, is one of the potential key factors in listeriosis treatment, coupled with the vast array of functions and strategies; this review summarizes the various strategies by LAB against L. monocytogenes and the prospect in development of a ‘generally regarded as safe’ LAB for treatment of listeriosis.
Collapse
|
11
|
The molecular mechanisms of listeriolysin O-induced lipid membrane damage. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183604. [PMID: 33722646 DOI: 10.1016/j.bbamem.2021.183604] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/22/2022]
Abstract
Listeria monocytogenes is an intracellular food-borne pathogen that causes listeriosis, a severe and potentially life-threatening disease. Listeria uses a number of virulence factors to proliferate and spread to various cells and tissues. In this process, three bacterial virulence factors, the pore-forming protein listeriolysin O and phospholipases PlcA and PlcB, play a crucial role. Listeriolysin O belongs to a family of cholesterol-dependent cytolysins that are mostly expressed by gram-positive bacteria. Its unique structural features in an otherwise conserved three-dimensional fold, such as the acidic triad and proline-glutamate-serine-threonine-like sequence, enable the regulation of its intracellular activity as well as distinct extracellular functions. The stability of listeriolysin O is pH- and temperature-dependent, and this provides another layer of control of its activity in cells. Moreover, many recent studies have demonstrated a unique mechanism of pore formation by listeriolysin O, i.e., the formation of arc-shaped oligomers that can subsequently fuse to form membrane defects of various shapes and sizes. During listerial invasion of host cells, these membrane defects can disrupt phagosome membranes, allowing bacteria to escape into the cytosol and rapidly multiply. The activity of listeriolysin O is profoundly dependent on the amount and accessibility of cholesterol in the lipid membrane, which can be modulated by the phospholipase PlcB. All these prominent features of listeriolysin O play a role during different stages of the L. monocytogenes life cycle by promoting the proliferation of the pathogen while mitigating excessive damage to its replicative niche in the cytosol of the host cell.
Collapse
|
12
|
Sharma A, Kanwar SS, Thakur SD. Biodiversity of meatborne Listeria spp. in Himachal Pradesh and their interaction with indigenous probiotics. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2021; 58:1209-1215. [PMID: 33678902 PMCID: PMC7884508 DOI: 10.1007/s13197-020-04854-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022]
Abstract
This study determined the anti-listerial activity of indigenous probiotics from traditional fermented foods of Western Himalaya against meat borne Listera monocytogens isolates from Himachal Pradesh. One hundred samples of meat and meat products like chicken (n = 25), chevon (goat meat, n = 20), fish (n = 20) and pork (n = 30) were collected and were analyzed for the presence of Listeria spp. by recommended culture and biochemical methods. L. monocytogens isolates were confirmed by PCR targeting the virulence gene hlyA (haemolysin A) and by16S rRNA sequencing. Anti-listerial activity of probiotic bacteria isolated from indigenous fermented foods of Himachal Pradesh was determined by well diffusion method using Lactobacillus rhamnosus GG (ATCC 53103) as the reference strain. Five percent of tested samples were found positive for L. monocytogens with incidence of 8.0% in chicken (2/25), 10.0% in fish (2/20) and 4.0% in chevon meat (1/25). None of the tested pork samples were found contaminated with L. monocytogenes. Among 11 indigenous probiotics used in this study, highest antagonistic activity was exhibited by Lactobacillus plantarum (ADF 10) and Enterococcus faecium (ADF1) which was equivalent to the reference strain.
Collapse
Affiliation(s)
- Aakriti Sharma
- Department of Microbiology, College of Basic Sciences, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh 176062 India
| | - S. S. Kanwar
- Department of Microbiology, College of Basic Sciences, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh 176062 India
| | - Sidharath Dev Thakur
- Department of Veterinary Public Health and Epidemiology, Dr. GC Negi College of Veterinary and Animal Sciences, CSK Himachal Pradesh Agricultural University, Palampur, Himachal Pradesh 176062 India
| |
Collapse
|
13
|
Phelps CC, Vadia S, Boyaka PN, Varikuti S, Attia Z, Dubey P, Satoskar AR, Tweten R, Seveau S. A listeriolysin O subunit vaccine is protective against Listeria monocytogenes. Vaccine 2020; 38:5803-5813. [PMID: 32684498 DOI: 10.1016/j.vaccine.2020.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 05/12/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022]
Abstract
Listeria monocytogenes is a facultative intracellular pathogen responsible for the life-threatening disease listeriosis. The pore-forming toxin listeriolysin O (LLO) is a critical virulence factor that plays a major role in the L. monocytogenes intracellular lifecycle and is indispensable for pathogenesis. LLO is also a dominant antigen for T cells involved in sterilizing immunity and it was proposed that LLO acts as a T cell adjuvant. In this work, we generated a novel full-length LLO toxoid (LLOT) in which the cholesterol-recognition motif, a threonine-leucine pair located at the tip of the LLO C-terminal domain, was substituted with two glycine residues. We showed that LLOT lost its ability to bind cholesterol and to form pores. Importantly, LLOT retained binding to the surface of epithelial cells and macrophages, suggesting that it could efficiently be captured by antigen-presenting cells. We then determined if LLOT can be used as an antigen and adjuvant to protect mice from L. monocytogenes infection. Mice were immunized with LLOT alone or together with cholera toxin or Alum as adjuvants. We found that mice immunized with LLOT alone or in combination with the Th2-inducing adjuvant Alum were not protected against L. monocytogenes. On the other hand, mice immunized with LLOT along with the experimental adjuvant cholera toxin, were protected against L. monocytogenes, as evidenced by a significant decrease in bacterial burden in the liver and spleen three days post-infection. This immunization regimen elicited mixed Th1, Th2, and Th17 responses, as well as the generation of LLO-neutralizing antibodies. Further, we identified T cells as being required for immunization-induced reductions in bacterial burden, whereas B cells were dispensable in our model of non-pregnant young mice. Overall, this work establishes that LLOT is a promising vaccine antigen for the induction of protective immunity against L. monocytogenes by subunit vaccines containing Th1-driving adjuvants.
Collapse
Affiliation(s)
- Christopher C Phelps
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA
| | - Stephen Vadia
- Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA; Department of Biology, Washington University in St. Louis, MO 63130, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Sanjay Varikuti
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Rodney Tweten
- Department of Microbial & Immunology, University of Oklahoma, Oklahoma City, OK, USA
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA; Department of Microbiology, The Ohio State University, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
14
|
Pouriayevali MH, Bamdad T, Sadat SM, Sadeghi SA, Sabahi F, Mahdavi M, Aghasadeghi MR. Listeriolysin O immunogenetic adjuvant enhanced potency of hepatitis C virus NS3 DNA vaccine. IUBMB Life 2019; 71:1645-1652. [PMID: 31298809 DOI: 10.1002/iub.2109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a major health problem all over the world. Among HCV proteins, nonstructural protein 3 (NS3) is one of the most promising target for anti-HCV therapy and a candidate for vaccine design. DNA vaccine is an efficient approach to stimulate antigen-specific immunity but the main problem with that is less immunogenic efficiency in comparison with traditional vaccines. Several approaches have been applied to enhance the immunogenicity of DNA. Recently, bacteria-derived substances are considered as one of the most attractive adjuvants for vaccines, which among them, Listeriolysin O (LLO) of Listeria monocytogenes is a toxin with an extremely immunogenic feature. We investigated detoxified form of LLO gene as genetic adjuvant to modulate NS3 DNA vaccine potency. Immunogenic truncated NS3 gene sequence of HCV (1095-1380aa) and detoxified LLO gene region (5-441aa) were amplified by PCR and cloned into the pcDNA3.1 plasmid separately. The expression of recombinant proteins (pc-NS3, pLLO) was confirmed in HEK293T cell line by western blotting. BALB/c mice models received three doses of different formula of plasmids in two-week intervals and two weeks after the final immunization, the immune responses were evaluated by specific total antibody level, lymphocyte proliferation, cytotoxicity, and cytokine levels assays. To evaluate in vivo cytotoxic activity, tumor challenge was performed. The recombinant plasmids were successfully expressed in mammalian cell line, and coadministration of pc-NS3 with pLLO induced the highest titer of total IgG against NS3 antigen compared with other controls. Determination of IgG subclasses confirmed the efficient increase in mixed responses with Th1 dominancy. Furthermore, significant levels of cytokines (p < .05) and lymphocyte proliferation responses (p < .05) indicated the superiority of this regimen. The findings may have important implication for LLO gene application as genetic adjuvant in immune response against HCV.
Collapse
Affiliation(s)
- Mohammad H Pouriayevali
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Taravat Bamdad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed M Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed A Sadeghi
- Department of Research and Development of Hepatitis A vaccine, Pasteur Institute of Iran, Alborz, Iran
| | - Farzaneh Sabahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
15
|
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
|
16
|
Vdovikova S, Luhr M, Szalai P, Nygård Skalman L, Francis MK, Lundmark R, Engedal N, Johansson J, Wai SN. A Novel Role of Listeria monocytogenes Membrane Vesicles in Inhibition of Autophagy and Cell Death. Front Cell Infect Microbiol 2017; 7:154. [PMID: 28516064 PMCID: PMC5413512 DOI: 10.3389/fcimb.2017.00154] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/12/2017] [Indexed: 11/13/2022] Open
Abstract
Bacterial membrane vesicle (MV) production has been mainly studied in Gram-negative species. In this study, we show that Listeria monocytogenes, a Gram-positive pathogen that causes the food-borne illness listeriosis, produces MVs both in vitro and in vivo. We found that a major virulence factor, the pore-forming hemolysin listeriolysin O (LLO), is tightly associated with the MVs, where it resides in an oxidized, inactive state. Previous studies have shown that LLO may induce cell death and autophagy. To monitor possible effects of LLO and MVs on autophagy, we performed assays for LC3 lipidation and LDH sequestration as well as analysis by confocal microscopy of HEK293 cells expressing GFP-LC3. The results revealed that MVs alone did not affect autophagy whereas they effectively abrogated autophagy induced by pure LLO or by another pore-forming toxin from Vibrio cholerae, VCC. Moreover, Listeria monocytogenes MVs significantly decreased Torin1-stimulated macroautophagy. In addition, MVs protected against necrosis of HEK293 cells caused by the lytic action of LLO. We explored the mechanisms of LLO-induced autophagy and cell death and demonstrated that the protective effect of MVs involves an inhibition of LLO-induced pore formation resulting in inhibition of autophagy and the lytic action on eukaryotic cells. Further, we determined that these MVs help bacteria to survive inside eukaryotic cells (mouse embryonic fibroblasts). Taken together, these findings suggest that intracellular release of MVs from L. monocytogenes may represent a bacterial strategy to survive inside host cells, by its control of LLO activity and by avoidance of destruction from the autophagy system during infection.
Collapse
Affiliation(s)
- Svitlana Vdovikova
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Morten Luhr
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Paula Szalai
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Lars Nygård Skalman
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Monika K Francis
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Richard Lundmark
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden.,Department of Integrative Medical Biology, Umeå UniversityUmeå, Sweden
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Jörgen Johansson
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Sun N Wai
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| |
Collapse
|
17
|
Wolf AJ, Liu GY, Underhill DM. Inflammatory properties of antibiotic-treated bacteria. J Leukoc Biol 2016; 101:127-134. [PMID: 27576461 DOI: 10.1189/jlb.4mr0316-153rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 12/12/2022] Open
Abstract
Antibiotics have proven to be enormously effective tools in combating infectious diseases. A common roadblock to the effective use of antibiotics is the development of antibiotic resistance. We have recently observed that the very mechanism by which methicillin-resistant Staphylococcus aureus (MRSA) becomes antibiotic resistant causes the organism to be more inflammatory to innate immune cells. In this review, we offer some thoughts on the ways in which antibiotics have been observed to influence immune responses to bacteria.
Collapse
Affiliation(s)
- Andrea J Wolf
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; and
| | - George Y Liu
- Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; and.,Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David M Underhill
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA; .,Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; and
| |
Collapse
|
18
|
Hashino M, Tachibana M, Nishida T, Hara H, Tsuchiya K, Mitsuyama M, Watanabe K, Shimizu T, Watarai M. Inactivation of the MAPK signaling pathway by Listeria monocytogenes infection promotes trophoblast giant cell death. Front Microbiol 2015; 6:1145. [PMID: 26528279 PMCID: PMC4607873 DOI: 10.3389/fmicb.2015.01145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022] Open
Abstract
Listeria monocytogenes has a well-characterized ability to cross the placental barrier, resulting in spontaneous abortion and fetal infections. However, the mechanisms resulting in infection-associated abortion are not fully understood. In this study, we demonstrate that the dephosphorylation of MAPK family proteins caused by L. monocytogenes infection of trophoblast giant (TG) cells, which are placental immune cells, contributes to infectious abortion. Dephosphorylation of c-Jun, p38, and ERK1/2 was observed in infected TG cells, causing the downregulation of cytoprotective heme oxygenase (HO)-1. Blocking the dephosphorylation of proteins, including MAPK family proteins, inhibited the decrease in HO-1 expression. Treatment with MAPK inhibitors inhibited bacterial internalization into TG cells. Moreover, Toll-like receptor 2 involved in the expression of MAPK family proteins. Infection with a listeriolysin O-deleted mutant impaired dephosphorylation of MAPK family proteins in TG cells and did not induce infectious abortion in a mouse model. These results suggest that inactivation of the MAPK pathway by L. monocytogenes induces TG cell death and causes infectious abortion.
Collapse
Affiliation(s)
- Masanori Hashino
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan
| | - Masato Tachibana
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan ; Division of Biomedical Food Research, National Institute of Health Sciences Tokyo, Japan
| | - Takashi Nishida
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan
| | - Hideki Hara
- Department of Microbiology, Graduate School of Medicine, Kyoto University Kyoto, Japan ; Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School Ann Arbor, MI, USA
| | - Kohsuke Tsuchiya
- Department of Microbiology, Graduate School of Medicine, Kyoto University Kyoto, Japan ; Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University Kanazawa, Japan
| | - Masao Mitsuyama
- Department of Microbiology, Graduate School of Medicine, Kyoto University Kyoto, Japan ; Graduate School of Advanced Integrated Studies in Human Survivability, Kyoto University Kyoto, Japan
| | - Kenta Watanabe
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan ; Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University at Yamaguchi Yamaguchi, Japan
| | - Takashi Shimizu
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan ; Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University at Yamaguchi Yamaguchi, Japan
| | - Masahisa Watarai
- The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida Campus Yamaguchi, Japan ; Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University at Yamaguchi Yamaguchi, Japan
| |
Collapse
|