1
|
García-Reyes RA, García-Cancino A, Arrevillaga-Boni G, Espinoza-Monje M, Gutiérrez-Zamorano C, Arrizon J, González-Avila M. Identification and Characterization of Probiotic Lactiplantibacillus plantarum BI-59.1 Isolated from tejuino and Its Capacity to Produce Biofilms. Curr Microbiol 2023; 80:220. [PMID: 37204589 DOI: 10.1007/s00284-023-03319-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 04/30/2023] [Indexed: 05/20/2023]
Abstract
Tejuino is a popular and traditional beverage consumed in north and western of Mexico, due to its biological properties, it is considered a natural source of probiotics. Nevertheless, few studies have been performed on Tejuino microbiota. In this work, the probiotic potential of the tejuino isolated Lactiplantibacillus plantarum BI-59.1 strain was investigated. Its effectiveness was compared with a commercial Lactobacillus spp and identified by 16S rDNA sequence homology. Lactiplantibacillus plantarum BI-59.1 strain showed probiotic properties, i.e., production of antimicrobial compounds (lactic acid and presence of plantaricin A gene), inhibition of entero-pathogens by planktonic cells and metabolites (Salmonella enterica serovar Typhimurium inhibition to HT29-MTX adhesion), biofilm formation, bacterial adhesion (HT29-MTX, 3.96 CFU/cell), and tolerance to stimulated gastrointestinal conditions (tolerance to pH 3 and bile salts). The strain was gamma hemolytic, susceptible to most antibiotics and negative for gelatinase production; thus, the Lactiplantibacillus. plantarum BI-59.1 strain is suitable for its use as a probiotic for nutraceutical or pharmaceutical formulations.
Collapse
Affiliation(s)
- Rudy Antonio García-Reyes
- Ex-Vivo Digestion Laboratory, Medical and Pharmaceutical Biotechnology Unit, Center for Research and Applied Technology in Jalisco (CIATEJ), Normalistas 800, Colinas de La Normal, 44270, Guadalajara, Jalisco, Mexico
| | - Apolinaria García-Cancino
- Laboratory of Bacterial Pathogenicity, Faculty of Biological Sciences, University of Concepcion, Chacabuco, 4030000, Concepción, Bío-Bío, Chile
| | - Gerardo Arrevillaga-Boni
- Ex-Vivo Digestion Laboratory, Medical and Pharmaceutical Biotechnology Unit, Center for Research and Applied Technology in Jalisco (CIATEJ), Normalistas 800, Colinas de La Normal, 44270, Guadalajara, Jalisco, Mexico
| | - Marcela Espinoza-Monje
- Laboratory of Bacterial Pathogenicity, Faculty of Biological Sciences, University of Concepcion, Chacabuco, 4030000, Concepción, Bío-Bío, Chile
| | - Cristian Gutiérrez-Zamorano
- Laboratory of Bacterial Pathogenicity, Faculty of Biological Sciences, University of Concepcion, Chacabuco, 4030000, Concepción, Bío-Bío, Chile
| | - Javier Arrizon
- Industrial Biotechnology Unit, Center for Research and Applied Technology in Jalisco (CIATEJ), Camino Arenero 1227, El Bajío del Arenal, 45019, Zapopan, Jalisco, Mexico
| | - Marisela González-Avila
- Ex-Vivo Digestion Laboratory, Medical and Pharmaceutical Biotechnology Unit, Center for Research and Applied Technology in Jalisco (CIATEJ), Normalistas 800, Colinas de La Normal, 44270, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
2
|
Limited Heme Oxygenase Contribution to Modulating the Severity of Salmonella enterica serovar Typhimurium Infection. Antioxidants (Basel) 2022; 11:antiox11061040. [PMID: 35739937 PMCID: PMC9219982 DOI: 10.3390/antiox11061040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 01/18/2023] Open
Abstract
An important virulence trait of Salmonella enterica serovar Typhimurium (S. Typhimurium) is the ability to avoid the host immune response, generating systemic and persistent infections. Host cells play a crucial role in bacterial clearance by expressing the enzyme heme oxygenase 1 (Hmox1), which catalyzes the degradation of heme groups into Fe2+, biliverdin, and carbon monoxide (CO). The role of Hmox1 activity during S. Typhimurium infection is not clear and previous studies have shown contradictory results. We evaluated the effect of pharmacologic modulation of Hmox1 in a mouse model of acute and persistent S. Typhimurium infection by administering the Hmox1 activity inductor cobalt protoporphyrin-IX (CoPP) or inhibitor tin protoporphyrin-IX (SnPP) before infection. To evaluate the molecular mechanism involved, we measured the colocalization of S. Typhimurium and autophagosome and lysosomal markers in macrophages. Administering CoPP reduced the bacterial burden in organs of mice 5 days post-infection, while SnPP-treated mice showed bacterial loads similar to vehicle-treated mice. Furthermore, CoPP reduced bacterial loads when administered after infection in macrophages in vitro and in a persistent infection model of S. Typhimurium in vivo, while tin protoporphyrin-IX (SnPP) treatment resulted in a bacterial burden similar to vehicle-treated controls. However, we did not observe significant differences in co-localization of green fluorescent protein (GFP)-labeled S. Typhimurium with the autophagic vesicles marker microtubule-associated protein 1A/1B-light chain 3 (LC3) and the lysosomal marker lysosomal-associated membrane protein 1 (LAMP-1) in macrophages treated with CoPP. Our results suggest that CoPP can enhance antimicrobial activity in response to Salmonella infection, reducing bacterial dissemination and persistence in mice, in a CO and autophagy- independent manner.
Collapse
|
3
|
Schultz BM, Melo-Gonzalez F, Salazar GA, Porto BN, Riedel CA, Kalergis AM, Bueno SM. New Insights on the Early Interaction Between Typhoid and Non-typhoid Salmonella Serovars and the Host Cells. Front Microbiol 2021; 12:647044. [PMID: 34276584 PMCID: PMC8282409 DOI: 10.3389/fmicb.2021.647044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica is a common source of food and water-borne infections, causing a wide range of clinical ailments in both human and animal hosts. Immunity to Salmonella involves an interplay between different immune responses, which are rapidly initiated to control bacterial burden. However, Salmonella has developed several strategies to evade and modulate the host immune responses. In this sense, the main knowledge about the pathogenicity of this bacterium has been obtained by the study of mouse models with non-typhoidal serovars. However, this knowledge is not representative of all the pathologies caused by non-typhoidal serovars in the human. Here we review the most important features of typhoidal and non-typhoidal serovars and the diseases they cause in the human host, describing the virulence mechanisms used by these pathogens that have been identified in different models of infection.
Collapse
Affiliation(s)
- Bárbara M Schultz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara N Porto
- Laboratory of Clinical and Experimental Immunology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.,Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Duan B, Shao L, Liu R, Msuthwana P, Hu J, Wang C. Lactobacillus rhamnosus GG defense against Salmonella enterica serovar Typhimurium infection through modulation of M1 macrophage polarization. Microb Pathog 2021; 156:104939. [PMID: 33964416 DOI: 10.1016/j.micpath.2021.104939] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/28/2023]
Abstract
Lactobacillus rhamnosus GG (LGG), a model probiotic strain, plays an important role in immune regulatory activity to prevent and treat intestinal inflammation or diarrhea. However, the effect of the immune modulation of LGG on macrophages to prevent Salmonella infection has not been thoroughly studied. In this study, C57BL/6 mice were pre-administered LGG for 7 days continuously, and then infected with Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium). The results of the in vivo study indicated that LGG could reduce body weight loss, death rate and intestinal inflammatory response caused by S. Typhimurium. LGG also limited S. Typhimurium dissemination to liver and spleen, and thereby protected against infection. In vitro study, we observed that LGG enhanced the phagocytic and bactericidal ability of macrophages and upregulated M1 macrophage characters (e.g. iNOS, NO and IL-12) against S. Typhimurium. In addition, LGG also elevated IL-10 secretion, which was helpful to ameliorate intestinal inflammatory injury caused by S. Typhimurium. In conclusion, LGG could modulate M1 macrophage polarization and offer protective effects against S. Typhimurium infection.
Collapse
Affiliation(s)
- Bingjie Duan
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Lina Shao
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Ruihan Liu
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Petunia Msuthwana
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Jingtao Hu
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China.
| |
Collapse
|
5
|
Interactions between Salmonella and host macrophages - Dissecting NF-κB signaling pathway responses. Microb Pathog 2021; 154:104846. [PMID: 33711426 DOI: 10.1016/j.micpath.2021.104846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/18/2020] [Accepted: 02/22/2021] [Indexed: 01/07/2023]
Abstract
Salmonella not only invades host cells, but also replicates intracellularly to cause a range of diseases, including gastroenteritis and systemic infections such as typhoid fever. The body's first line of defense against pathogens is the innate immune response system that can protect against Salmonella invasion and replication. Nuclear factor κB (NF-κB) is an important transcriptional regulator that plays an important role in host inflammatory responses to pathogens. Both the canonical and non-canonical NF-κB signaling pathways are activated by Salmonella in many different ways through its virulence factors, leading to the release of inflammatory factors and the activation of inflammatory responses in mammalian hosts. Equally, Salmonella, as an enteropathogen, has accordingly evolved strategies to disturb NF-κB activation, such as secreting some effector proteins by type III secretion systems as well as inducing host cells to express NF-κB pathway inhibitors, allowing it to colonize and persistently infect the hosts. This review focuses on how Salmonella activates NF-κB signaling pathway and the strategies used by Salmonella to interfere with the NF-κB pathway activation.
Collapse
|
6
|
Loh FK, Nathan S, Chow SC, Fang CM. Immunogenicity and protection efficacy of enhanced fitness recombinant Salmonella Typhi monovalent and bivalent vaccine strains against acute toxoplasmosis. Pathog Glob Health 2021; 115:392-404. [PMID: 33525974 DOI: 10.1080/20477724.2021.1881369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The proficiency of Salmonella Typhi to induce cell-mediated immunity has allowed its exploitation as a live vector against the obligate intracellular protozoan Toxoplasma gondii. T. gondii vaccine research is of great medical value due to the lack of a suitable toxoplasmosis vaccine. In the present work, we integrated T. gondii antigen into a growth-dependent chromosome locus guaBA of S. Typhi CVD910 strain to form recombinant S. Typhi monovalent CVD910-SAG1 expressed T. gondii SAG1 antigen and monovalent CVD910-GRA2 expressed T. gondii GRA2 antigen. Furthermore, a low-copy stabilized recombinant plasmid encoding SAG1 antigen was transformed into CVD910-GRA2 to form bivalent CVD910-GS strain. An osmolarity-regulated promoter was also incorporated to control the gene transcription, whereas clyA export protein was included to translocate the antigen out of the cytoplasm. Both CVD910-GRA2 and CVD910-GS displayed healthy growth fitness and readily expressed the encoded T. gondii antigens. When administered in vivo, CVD910-GS successfully induced both humoral and cellular immunity in the immunized BALB/c mice, and extended mice survival against virulent T. gondii. In particular, the mice immunized with bivalent CVD910-GS presented the highest titers of IgG, percentages of CD4+ T, CD8+ T, B cells and memory T cells, and total IgG+ memory B cells as compared to the CVD910-GRA2 and control strains. The CVD910-GS group also generated mixed Th1/Th2 cytokine profile with secretions of IFN-ɣ, IL-2 and IL-10. This study demonstrated the importance of enhancing live vector fitness to sustain heterologous antigen expression for eliciting robust immune responses and providing effective protection against pathogen.
Collapse
Affiliation(s)
- Fei-Kean Loh
- Division of Biomedical Sciences, School of Pharmacy, the University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Sheila Nathan
- Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, the University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| |
Collapse
|
7
|
Xiong D, Song L, Geng S, Jiao Y, Zhou X, Song H, Kang X, Zhou Y, Xu X, Sun J, Pan Z, Jiao X. Salmonella Coiled-Coil- and TIR-Containing TcpS Evades the Innate Immune System and Subdues Inflammation. Cell Rep 2020; 28:804-818.e7. [PMID: 31315056 DOI: 10.1016/j.celrep.2019.06.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/23/2019] [Accepted: 06/13/2019] [Indexed: 01/12/2023] Open
Abstract
Toll-like receptors (TLRs) activate innate immunity via interactions between their Toll/interleukin-1 (IL-1) receptor (TIR) domain and downstream adaptor proteins. Here we report that Salmonella Enteritidis produces a secreted protein (TcpS) that contains both a TIR domain and a coiled-coil domain. TcpS blocks MyD88- and TRIF-mediated TLR signaling, inhibits inflammatory responses, and promotes bacterial survival. Early-stage immune evasion by TcpS results in severe tissue damage in the late stage of infection and contributes to Salmonella virulence. TcpS-derived peptides inhibit nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) activation and reduce lipopolysaccharide (LPS)-elicited systemic inflammation. Therapeutic peptide administration alleviates weight loss of mice infected with H1N1 influenza. Importantly, maximal TcpS-mediated TLR inhibition requires the critical TIR-TcpS residues Y191 and I284, as well as TcpS homodimerization via its N-terminal coiled-coil domain. Our study unveils a mechanism in which TcpS suppresses innate immunity via both its homodimerization and interaction with MyD88. TcpS is also a potential therapeutic agent for inflammation-associated diseases.
Collapse
Affiliation(s)
- Dan Xiong
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Li Song
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yang Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiaohui Zhou
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Hongqin Song
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xilong Kang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiulong Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
8
|
Häfner S. Conquest of Paratyphi. Microbes Infect 2019; 22:285-289. [PMID: 31838130 DOI: 10.1016/j.micinf.2019.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Sophia Häfner
- University of Copenhagen, BRIC Biotech Research & Innovation Centre, Lund Group, 2200 Copenhagen, Denmark.
| |
Collapse
|
9
|
|
10
|
Aulicino A, Rue-Albrecht KC, Preciado-Llanes L, Napolitani G, Ashley N, Cribbs A, Koth J, Lagerholm BC, Ambrose T, Gordon MA, Sims D, Simmons A. Invasive Salmonella exploits divergent immune evasion strategies in infected and bystander dendritic cell subsets. Nat Commun 2018; 9:4883. [PMID: 30451854 PMCID: PMC6242960 DOI: 10.1038/s41467-018-07329-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/25/2018] [Indexed: 01/06/2023] Open
Abstract
Non-typhoidal Salmonella (NTS) are highly prevalent food-borne pathogens. Recently, a highly invasive, multi-drug resistant S. Typhimurium, ST313, emerged as a major cause of bacteraemia in children and immunosuppressed adults, however the pathogenic mechanisms remain unclear. Here, we utilize invasive and non-invasive Salmonella strains combined with single-cell RNA-sequencing to study the transcriptome of individual infected and bystander monocyte-derived dendritic cells (MoDCs) implicated in disseminating invasive ST313. Compared with non-invasive Salmonella, ST313 directs a highly heterogeneous innate immune response. Bystander MoDCs exhibit a hyper-activated profile potentially diverting adaptive immunity away from infected cells. MoDCs harbouring invasive Salmonella display higher expression of IL10 and MARCH1 concomitant with lower expression of CD83 to evade adaptive immune detection. Finally, we demonstrate how these mechanisms conjointly restrain MoDC-mediated activation of Salmonella-specific CD4+ T cell clones. Here, we show how invasive ST313 exploits discrete evasion strategies within infected and bystander MoDCs to mediate its dissemination in vivo.
Collapse
Affiliation(s)
- Anna Aulicino
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Kevin C Rue-Albrecht
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7FY, UK
| | - Lorena Preciado-Llanes
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Giorgio Napolitani
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Neil Ashley
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford and BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford, OX3 9DS, UK
| | - Adam Cribbs
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Jana Koth
- MRC Human Immunology Unit and Wolfson Imaging Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - B Christoffer Lagerholm
- MRC Human Immunology Unit and Wolfson Imaging Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Tim Ambrose
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Melita A Gordon
- Institute of Infection and Global Health, University of Liverpool, 8 W Derby St, Liverpool, L7 3EA, UK
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - David Sims
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Alison Simmons
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| |
Collapse
|
11
|
Letendre C, Auger JP, Lemire P, Galbas T, Gottschalk M, Thibodeau J, Segura M. Streptococcus suis Serotype 2 Infection Impairs Interleukin-12 Production and the MHC-II-Restricted Antigen Presentation Capacity of Dendritic Cells. Front Immunol 2018; 9:1199. [PMID: 29899744 PMCID: PMC5988873 DOI: 10.3389/fimmu.2018.01199] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/14/2018] [Indexed: 01/18/2023] Open
Abstract
Streptococcus suis is an important swine pathogen and emerging zoonotic agent. Encapsulated strains of S. suis modulate dendritic cell (DC) functions, leading to poorly activated CD4+ T cells. However, the antigen presentation ability of S. suis-stimulated DCs has not been investigated yet. In this work, we aimed to characterize the antigen presentation profiles of S. suis-stimulated DCs, both in vitro and in vivo. Upon direct activation in vitro, S. suis-stimulated murine bone marrow-derived DCs (bmDCs) preserved their antigen capture/processing capacities. However, they showed delayed kinetics of MHC-II expression compared to lipopolysaccharide-stimulated bmDCs. Meanwhile, splenic DCs from infected mice exhibited a compromised MHC-II expression, despite an appropriate expression of maturation markers. To identify potential interfering mechanisms, Class II Major Histocompatibility Complex Transactivator (CIITA) and membrane-associated RING-CH (MARCH)1/8 transcription were studied. S. suis-stimulated DCs maintained low levels of CIITA at early time points, both in vitro and in vivo, which could limit their ability to increase MHC-II synthesis. S. suis-stimulated DCs also displayed sustained/upregulated levels of MARCH1/8, thus possibly leading to MHC-II lysosomal degradation. The bacterial capsular polysaccharide played a partial role in this modulation. Finally, interleukin (IL)-12p70 production was inhibited in splenic DCs from infected mice, a profile compatible with DC indirect activation by pro-inflammatory compounds. Consequently, these cells induced lower levels of IL-2 and TNF-α in an antigen-specific CD4+ T cell presentation assay and blunted T cell CD25 expression. It remains unclear at this stage whether these phenotypical and transcriptional modulations observed in response to S. suis in in vivo infections are part of a bacterial immune evasion strategy or rather a feature common to systemic inflammatory response-inducing agents. However, it appears that the MHC-II-restricted antigen presentation and Th1-polarizing cytokine production capacities of DCs are impaired during S. suis infection. This study highlights the potential consequences of inflammation on the type and magnitude of the immune response elicited by a pathogen.
Collapse
Affiliation(s)
- Corinne Letendre
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Jean-Philippe Auger
- Laboratory of Research on Streptococcus suis, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Paul Lemire
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Tristan Galbas
- Laboratory of Molecular Immunology, Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada
| | - Marcelo Gottschalk
- Laboratory of Research on Streptococcus suis, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Jacques Thibodeau
- Laboratory of Molecular Immunology, Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada
| | - Mariela Segura
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| |
Collapse
|
12
|
dos Santos AMP, Ferrari RG, Conte-Junior CA. Virulence Factors in Salmonella Typhimurium: The Sagacity of a Bacterium. Curr Microbiol 2018; 76:762-773. [DOI: 10.1007/s00284-018-1510-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 05/16/2018] [Indexed: 12/20/2022]
|
13
|
Rey-Jurado E, Tapia F, Muñoz-Durango N, Lay MK, Carreño LJ, Riedel CA, Bueno SM, Genzel Y, Kalergis AM. Assessing the Importance of Domestic Vaccine Manufacturing Centers: An Overview of Immunization Programs, Vaccine Manufacture, and Distribution. Front Immunol 2018; 9:26. [PMID: 29403503 PMCID: PMC5778105 DOI: 10.3389/fimmu.2018.00026] [Citation(s) in RCA: 11] [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: 11/11/2017] [Accepted: 01/04/2018] [Indexed: 12/03/2022] Open
Abstract
Vaccines have significantly reduced the detrimental effects of numerous human infectious diseases worldwide, helped to reduce drastically child mortality rates and even achieved eradication of major pathogens, such as smallpox. These achievements have been possible due to a dedicated effort for vaccine research and development, as well as an effective transfer of these vaccines to public health care systems globally. Either public or private institutions have committed to developing and manufacturing vaccines for local or international population supply. However, current vaccine manufacturers worldwide might not be able to guarantee sufficient vaccine supplies for all nations when epidemics or pandemics events could take place. Currently, different countries produce their own vaccine supplies under Good Manufacturing Practices, which include the USA, Canada, China, India, some nations in Europe and South America, such as Germany, the Netherlands, Italy, France, Argentina, and Brazil, respectively. Here, we discuss some of the vaccine programs and manufacturing capacities, comparing the current models of vaccine management between industrialized and developing countries. Because local vaccine production undoubtedly provides significant benefits for the respective population, the manufacture capacity of these prophylactic products should be included in every country as a matter of national safety.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Tapia
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
14
|
Retamal-Díaz AR, Kalergis AM, Bueno SM, González PA. A Herpes Simplex Virus Type 2 Deleted for Glycoprotein D Enables Dendritic Cells to Activate CD4 + and CD8 + T Cells. Front Immunol 2017; 8:904. [PMID: 28848543 PMCID: PMC5553038 DOI: 10.3389/fimmu.2017.00904] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/14/2017] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus type 2 (HSV-2) is highly prevalent in the human population producing significant morbidity, mainly because of the generation of genital ulcers and neonatal encephalitis. Additionally, HSV-2 infection significantly increases the susceptibility of the host to acquire HIV and promotes the shedding of the latter in the coinfected. Despite numerous efforts to create a vaccine against HSV-2, no licensed vaccines are currently available. A long-standing strategy, based on few viral glycoproteins combined with adjuvants, recently displayed poor results in a Phase III clinical study fueling exploration on the development of mutant HSV viruses that are attenuated in vivo and elicit protective adaptive immune components, such as antiviral antibodies and T cells. Importantly, such specialized antiviral immune components are likely induced and modulated by dendritic cells, professional antigen presenting cells that process viral antigens and present them to T cells. However, HSV interferes with several functions of DCs and ultimately induces their death. Here, we propose that for an attenuated mutant virus to confer protective immunity against HSV in vivo based on adaptive immune components, such virus should also be attenuated in dendritic cells to promote a robust and effective antiviral response. We provide a background framework for this idea, considerations, as well as the means to assess this hypothesis. Addressing this hypothesis may provide valuable insights for the development of novel, safe, and effective vaccines against herpes simplex viruses.
Collapse
Affiliation(s)
- Angello R Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,INSERM U1064, Nantes, France
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Wagner RD, Johnson SJ. Probiotic bacteria prevent Salmonella - induced suppression of lymphoproliferation in mice by an immunomodulatory mechanism. BMC Microbiol 2017; 17:77. [PMID: 28356067 PMCID: PMC5372341 DOI: 10.1186/s12866-017-0990-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/23/2017] [Indexed: 11/25/2022] Open
Abstract
Background Salmonella enterica infections often exhibit a form of immune evasion. We previously observed that probiotic bacteria could prevent inhibition of lymphoproliferation and apoptosis responses of T cells associated with S. enterica infections in orally challenged mice. Results In this study, changes in expression of genes related to lymphocyte activation in mucosa-associated lymphoid tissues (MALT) of mice orally infected with S. enterica with and without treatment with probiotic bacteria were evaluated. Probiotic bacteria increased expression of mRNA for clusters of differentiation antigen 2 (Cd2), protein tyrosine phosphatase receptor type C (Ptprc), and Toll-like receptor 6 (Tlr6) genes related to T and B cell activation in mouse intestinal tissue. The probiotic bacteria were also associated with reduced mRNA expression of a group of genes (RelB, Myd88, Iκκa, Jun, Irak2) related to nuclear factor of kappa light chains enhancer in B cells (NF-κB) signal transduction pathway-regulated cytokine responses. Probiotic bacteria were also associated with reduced mRNA expression of apoptotic genes (Casp2, Casp12, Dad1, Akt1, Bad) that suggest high avidity lymphocyte sparing. Reduced CD2 immunostaining in mesenteric lymph nodes (MLN) was suggestive of reduced lymphocyte activation in probiotic-treated mice. Reduced immunostaining of TLR6 in MALT of probiotic-treated, S. enterica-infected mice suggests that diminished innate immune sensitivity to S. enterica antigens is associated with preventing lymphocyte deletion. Conclusions The results of this study are consistent with prevention of S. enterica-induced deletion of lymphocytes by the influence of probiotic bacteria in mucosal lymphoid tissues of mice.
Collapse
Affiliation(s)
- R Doug Wagner
- Microbiology Division, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| | - Shemedia J Johnson
- Microbiology Division, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| |
Collapse
|
16
|
Schultz BM, Paduro CA, Salazar GA, Salazar-Echegarai FJ, Sebastián VP, Riedel CA, Kalergis AM, Alvarez-Lobos M, Bueno SM. A Potential Role of Salmonella Infection in the Onset of Inflammatory Bowel Diseases. Front Immunol 2017; 8:191. [PMID: 28293241 PMCID: PMC5329042 DOI: 10.3389/fimmu.2017.00191] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/09/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) includes a set of pathologies that result from a deregulated immune response that may affect any portion of the gastrointestinal tract. The most prevalent and defined forms of IBD are Crohn’s disease and ulcerative colitis. Although the etiology of IBD is not well defined, it has been suggested that environmental and genetic factors contribute to disease development and that the interaction between these two factors can trigger the pathology. Diet, medication use, vitamin D status, smoking, and bacterial infections have been proposed to influence or contribute to the onset or development of the disease in susceptible individuals. The infection with pathogenic bacteria is a key factor that can influence the development and severity of this disease. Here, we present a comprehensive review of studies performed in human and mice susceptible to IBD, which supports the notion that infection with bacterial pathogens, such as Salmonella, could promote the onset of IBD due to permanent changes in the intestinal microbiota, disruption of the epithelial barrier and alterations of the intestinal immune response after infection.
Collapse
Affiliation(s)
- Bárbara M Schultz
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Carolina A Paduro
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Geraldyne A Salazar
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Francisco J Salazar-Echegarai
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Valentina P Sebastián
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Claudia A Riedel
- Facultad de Ciencias Biológicas y Facultad de Medicina, Departamento de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello , Santiago , Chile
| | - Alexis M Kalergis
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile; Facultad de Medicina, Departamento de Endocrinología, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM, UMR 1064, Nantes, France
| | - Manuel Alvarez-Lobos
- Facultad de Medicina, Departamento de Gastroenterología, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Susan M Bueno
- Facultad de Ciencias Biológicas, Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM, UMR 1064, Nantes, France
| |
Collapse
|
17
|
McLaughlin PA, McClelland M, Yang HJ, Porwollik S, Bogomolnaya L, Chen JS, Andrews-Polymenis H, van der Velden AWM. Contribution of Asparagine Catabolism to Salmonella Virulence. Infect Immun 2017; 85:e00740-16. [PMID: 27849183 PMCID: PMC5278173 DOI: 10.1128/iai.00740-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/10/2016] [Indexed: 02/01/2023] Open
Abstract
Salmonellae are pathogenic bacteria that cause significant morbidity and mortality in humans worldwide. Salmonellae establish infection and avoid clearance by the immune system by mechanisms that are not well understood. We previously showed that l-asparaginase II produced by Salmonella enterica serovar Typhimurium (S Typhimurium) inhibits T cell responses and mediates virulence. In addition, we previously showed that asparagine deprivation such as that mediated by l-asparaginase II of S Typhimurium causes suppression of activation-induced T cell metabolic reprogramming. Here, we report that STM3997, which encodes a homolog of disulfide bond protein A (dsbA) of Escherichia coli, is required for l-asparaginase II stability and function. Furthermore, we report that l-asparaginase II localizes primarily to the periplasm and acts together with l-asparaginase I to provide S Typhimurium the ability to catabolize asparagine and assimilate nitrogen. Importantly, we determined that, in a murine model of infection, S Typhimurium lacking both l-asparaginase I and II genes competes poorly with wild-type S Typhimurium for colonization of target tissues. Collectively, these results indicate that asparagine catabolism contributes to S Typhimurium virulence, providing new insights into the competition for nutrients at the host-pathogen interface.
Collapse
Affiliation(s)
- Patrick A McLaughlin
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Hee-Jeong Yang
- Department of Microbial Pathogenesis and Immunology, Texas A&M University System Health Science Center, Bryan, Texas, USA
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Lydia Bogomolnaya
- Department of Microbial Pathogenesis and Immunology, Texas A&M University System Health Science Center, Bryan, Texas, USA
| | - Juei-Suei Chen
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Helene Andrews-Polymenis
- Department of Microbial Pathogenesis and Immunology, Texas A&M University System Health Science Center, Bryan, Texas, USA
| | - Adrianus W M van der Velden
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
18
|
Wagnerova A, Babickova J, Liptak R, Celec P, Gardlik R. Beneficial effects of live and dead Salmonella
-based vector strain on the course of colitis in mice. Lett Appl Microbiol 2016; 63:340-346. [DOI: 10.1111/lam.12632] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022]
Affiliation(s)
- A. Wagnerova
- Institute of Molecular Biomedicine; Faculty of Medicine; Comenius University; Bratislava Slovakia
| | - J. Babickova
- Institute of Molecular Biomedicine; Faculty of Medicine; Comenius University; Bratislava Slovakia
- Institute for Clinical and Translational Research BMC; Biomedical Research Center; Slovak Academy of Sciences; Bratislava Slovakia
| | - R. Liptak
- Institute of Molecular Biomedicine; Faculty of Medicine; Comenius University; Bratislava Slovakia
| | - P. Celec
- Institute of Molecular Biomedicine; Faculty of Medicine; Comenius University; Bratislava Slovakia
- Institute for Clinical and Translational Research BMC; Biomedical Research Center; Slovak Academy of Sciences; Bratislava Slovakia
- Institute of Pathophysiology; Faculty of Medicine; Comenius University; Bratislava Slovakia
- Department of Molecular Biology; Faculty of Natural Sciences; Comenius University; Bratislava Slovakia
| | - R. Gardlik
- Institute of Molecular Biomedicine; Faculty of Medicine; Comenius University; Bratislava Slovakia
- Institute for Clinical and Translational Research BMC; Biomedical Research Center; Slovak Academy of Sciences; Bratislava Slovakia
- Institute of Pathophysiology; Faculty of Medicine; Comenius University; Bratislava Slovakia
| |
Collapse
|
19
|
Peñaloza HF, Schultz BM, Nieto PA, Salazar GA, Suazo I, Gonzalez PA, Riedel CA, Alvarez-Lobos MM, Kalergis AM, Bueno SM. Opposing roles of IL-10 in acute bacterial infection. Cytokine Growth Factor Rev 2016; 32:17-30. [PMID: 27522641 DOI: 10.1016/j.cytogfr.2016.07.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Interleukin-10 (IL-10) is recognized as an anti-inflammatory cytokine that downmodulates inflammatory immune responses at multiple levels. In innate cells, production of this cytokine is usually triggered after pathogen recognition receptor (PRR) engagement by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patters (DAMPs), as well as by other soluble factors. Importantly, IL-10 is frequently secreted during acute bacterial infections and has been described to play a key role in infection resolution, although its effects can significantly vary depending on the infecting bacterium. While the production of IL-10 might favor host survival in some cases, it may also result harmful for the host in other circumstances, as it can prevent appropriate bacterial clearance. In this review we discuss the role of IL-10 in bacterial clearance and propose that this cytokine is required to recover from infection caused by extracellular or highly pro-inflammatory bacteria. Altogether, we propose that IL-10 drives excessive suppression of the immune response upon infection with intracellular bacteria or in non-inflammatory bacterial infections, which ultimately favors bacterial persistence and dissemination within the host. Thus, the nature of the bacterium causing infection is an important factor that needs to be taken into account when considering new immunotherapies that consist on the modulation of inflammation, such as IL-10. Indeed, induction of this cytokine may significantly improve the host's immune response to certain bacteria when antibiotics are not completely effective.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Barbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Geraldyne A Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Isidora Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pablo A Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Chile
| | - Manuel M Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
20
|
Torres A, Luke JD, Kullas AL, Kapilashrami K, Botbol Y, Koller A, Tonge PJ, Chen EI, Macian F, van der Velden AWM. Asparagine deprivation mediated by Salmonella asparaginase causes suppression of activation-induced T cell metabolic reprogramming. J Leukoc Biol 2015; 99:387-98. [PMID: 26497246 DOI: 10.1189/jlb.4a0615-252r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/02/2015] [Indexed: 01/09/2023] Open
Abstract
Salmonellae are pathogenic bacteria that induce immunosuppression by mechanisms that remain largely unknown. Previously, we showed that a putative type II l-asparaginase produced by Salmonella Typhimurium inhibits T cell responses and mediates virulence in a murine model of infection. Here, we report that this putative L-asparaginase exhibits L-asparagine hydrolase activity required for Salmonella Typhimurium to inhibit T cells. We show that L-asparagine is a nutrient important for T cell activation and that L-asparagine deprivation, such as that mediated by the Salmonella Typhimurium L-asparaginase, causes suppression of activation-induced mammalian target of rapamycin signaling, autophagy, Myc expression, and L-lactate secretion. We also show that L-asparagine deprivation mediated by the Salmonella Typhimurium L-asparaginase causes suppression of cellular processes and pathways involved in protein synthesis, metabolism, and immune response. Our results advance knowledge of a mechanism used by Salmonella Typhimurium to inhibit T cell responses and mediate virulence, and provide new insights into the prerequisites of T cell activation. We propose a model in which l-asparagine deprivation inhibits T cell exit from quiescence by causing suppression of activation-induced metabolic reprogramming.
Collapse
Affiliation(s)
- AnnMarie Torres
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Joanna D Luke
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amy L Kullas
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kanishk Kapilashrami
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yair Botbol
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Antonius Koller
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Peter J Tonge
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Emily I Chen
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Fernando Macian
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Adrianus W M van der Velden
- *Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Graduate Program in Genetics, Department of Chemistry and Institute for Chemical Biology and Drug Discovery, Proteomics Center, and Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; and Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
21
|
Kramer MG, Masner M, Casales E, Moreno M, Smerdou C, Chabalgoity JA. Neoadjuvant administration of Semliki Forest virus expressing interleukin-12 combined with attenuated Salmonella eradicates breast cancer metastasis and achieves long-term survival in immunocompetent mice. BMC Cancer 2015; 15:620. [PMID: 26347489 PMCID: PMC4562361 DOI: 10.1186/s12885-015-1618-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metastatic breast cancer is a major cause of death among women worldwide; therefore efficient therapeutic strategies are extremely needed. In this work we have developed a gene therapy- and bacteria-based combined neoadjuvant approach and evaluated its antitumor effect in a clinically relevant animal model of metastatic breast cancer. METHODS 2×10(8) particles of a Semliki Forest virus vector expressing interleukin-12 (SFV-IL-12) and/or 2×10(7) units of an aroC (-) Samonella Typhimurium strain (LVR01) were injected into 4T1 tumor nodules orthotopically implanted in mice. Tumors were surgically resected and long-term survival was determined. IL-12 and interferon-γ were quantified by Enzyme-Linked ImmunoSorbent Assay, bacteria was visualized by inmunohistochemistry and the number of lung metastasis was calculated with a clonogenic assay. RESULTS SFV-IL-12 and LVR01 timely inoculated and followed by surgical resection of tumors succeeded in complete inhibition of lethal lung metastasis and long-term survival in 90% of treated mice. The combined therapy was markedly synergistic compared to each treatment alone, since SFV-IL-12 monotherapy showed a potent antiangiogenic effect, being able to inhibit tumor growth and extend survival, but could not prevent establishment of distant metastasis and death of tumor-excised animals. On the other hand, LVR01 alone also showed a significant, although limited, antitumor potential, despite its ability to invade breast cancer cells and induce granulocyte recruitment. The efficacy of the combined therapy depended on the order in which both factors were administered; inasmuch the therapeutic effect was only observed when SFV-IL-12 was administered previous to LVR01, whereas administration of LVR01 before SFV-IL-12 had negligible antitumor activity. Moreover, pre-treatment with LVR01 seemed to suppress SFV-IL-12 antiangiogenic effects associated to lower IL-12 expression in this group. Re-challenged mice were unable to reject a second 4T1 tumor; however 100% of them could be totally cured by applying the same neoadjuvant combined regimen. To our knowledge, these are the most encouraging results obtained to date in a post-operatory setting using the highly aggressive 4T1 animal model. CONCLUSIONS SFV-IL-12-based gene therapy combined with Salmonella LVR01 neoadjuvant administration has a synergic antitumor effect and may be a promising therapeutic option to prevent and/or eradicate pre-operatory metastasis in locally advanced breast cancer.
Collapse
Affiliation(s)
- M Gabriela Kramer
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Martín Masner
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Erkuden Casales
- Division Gene Therapy, Center for Applied Medical Research, University of Navarra, Av. Pio XII 55, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, c/Irunlarrea 3, 31008, Pamplona, Spain.
| | - María Moreno
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| | - Cristian Smerdou
- Division Gene Therapy, Center for Applied Medical Research, University of Navarra, Av. Pio XII 55, 31008, Pamplona, Spain.
- IdiSNA, Navarra Institute for Health Research, c/Irunlarrea 3, 31008, Pamplona, Spain.
| | - José A Chabalgoity
- Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, (UdelaR), Av. A. Navarro 3051, 11600, Montevideo, Uruguay.
| |
Collapse
|
22
|
Roscetto E, Vitiello L, Muoio R, Soriano AA, Iula VD, Vollaro A, De Gregorio E, Catania MR. In vitro interaction of Stenotrophomonas maltophilia with human monocyte-derived dendritic cells. Front Microbiol 2015; 6:723. [PMID: 26236302 PMCID: PMC4504169 DOI: 10.3389/fmicb.2015.00723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/02/2015] [Indexed: 11/20/2022] Open
Abstract
Stenotrophomonas maltophilia is increasingly identified as an opportunistic pathogen in immunocompromised, cancer and cystic fibrosis (CF) patients. Knowledge on innate immune responses to S. maltophilia and its potential modulation is poor. The present work investigated the ability of 12 clinical S. maltophilia strains (five from CF patients, seven from non-CF patients) and one environmental strain to survive inside human monocyte-derived dendritic cells (DCs). The effects of the bacteria on maturation of and cytokine secretion by DCs were also measured. S. maltophilia strains presented a high degree of heterogeneity in internalization and intracellular replication efficiencies as well as in the ability of S. maltophilia to interfere with normal DCs maturation. By contrast, all S. maltophilia strains were able to activate DCs, as measured by increase in the expression of surface maturation markers and proinflammatory cytokines secretion.
Collapse
Affiliation(s)
- Emanuela Roscetto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| | - Laura Vitiello
- Laboratorio di Immunologia Cellulare e Molecolare, Istituto di Ricovero e Cura a Carattere Scientifico , San Raffaele Pisana, Rome, Italy
| | - Rosa Muoio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| | - Amata A Soriano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| | - Vita D Iula
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| | - Antonio Vollaro
- DAI Medicina di Laboratorio, Azienda Ospedaliera Universitaria Federico II , Napoli, Italy
| | - Eliana De Gregorio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| | - Maria R Catania
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II , Napoli, Italy
| |
Collapse
|
23
|
Verma S, Srikanth CV. Understanding the complexities of Salmonella-host crosstalk as revealed by in vivo model organisms. IUBMB Life 2015; 67:482-97. [PMID: 26179888 DOI: 10.1002/iub.1393] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 01/03/2023]
Abstract
Foodborne infections caused by non-typhoidal Salmonellae, such as Salmonella enterica serovar Typhimurium (ST), pose a major challenge in the developed and developing world. With constant rise of drug-resistant strains, understanding the epidemiology, microbiology, pathogenesis and host-pathogen interactions biology is a mandatory requirement to enable health systems to be ready to combat these illnesses. Patient data from hospitals, at least from some parts of the world, have aided in epidemiological understanding of ST-mediated disease. Most of the other aspects connected to Salmonella-host crosstalk have come from model systems that offer convenience, genetic tractability and low maintenance costs that make them extremely valuable tools. Complex model systems such as the bovine model have helped in understanding key virulence factors needed for infection. Simple systems such as fruit flies and Caenorhabditis elegans have aided in identification of novel virulence factors, host pathways and mechanistic details of interactions. Some of the path-breaking concepts of the field have come from mice model of ST colitis, which allows genetic manipulations as well as high degree of similarity to human counterpart. Together, they are invaluable for correlating in vitro findings of ST-induced disease progression in vivo. The current review is a compilation of various advances of ST-host interactions at cellular and molecular levels that has come from investigations involving model organisms.
Collapse
Affiliation(s)
- Smriti Verma
- Regional Centre for Biotechnology, NCR Biotech Cluster 3rd Milestone, Gurgaon-Faridabad Highway, Village Bhankari, Faridabad, Haryana, India
| | - Chittur V Srikanth
- Regional Centre for Biotechnology, NCR Biotech Cluster 3rd Milestone, Gurgaon-Faridabad Highway, Village Bhankari, Faridabad, Haryana, India
| |
Collapse
|
24
|
Banerji J. Asparaginase treatment side-effects may be due to genes with homopolymeric Asn codons (Review-Hypothesis). Int J Mol Med 2015; 36:607-26. [PMID: 26178806 PMCID: PMC4533780 DOI: 10.3892/ijmm.2015.2285] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/15/2015] [Indexed: 12/14/2022] Open
Abstract
The present treatment of childhood T-cell leukemias involves the systemic administration of prokary-otic L-asparaginase (ASNase), which depletes plasma Asparagine (Asn) and inhibits protein synthesis. The mechanism of therapeutic action of ASNase is poorly understood, as are the etiologies of the side-effects incurred by treatment. Protein expression from genes bearing Asn homopolymeric coding regions (N-hCR) may be particularly susceptible to Asn level fluctuation. In mammals, N-hCR are rare, short and conserved. In humans, misfunctions of genes encoding N-hCR are associated with a cluster of disorders that mimic ASNase therapy side-effects which include impaired glycemic control, dislipidemia, pancreatitis, compromised vascular integrity, and neurological dysfunction. This paper proposes that dysregulation of Asn homeostasis, potentially even by ASNase produced by the microbiome, may contribute to several clinically important syndromes by altering expression of N-hCR bearing genes. By altering amino acid abundance and modulating ribosome translocation rates at codon repeats, the microbiomic environment may contribute to genome decoding and to shaping the proteome. We suggest that impaired translation at poly Asn codons elevates diabetes risk and severity.
Collapse
Affiliation(s)
- Julian Banerji
- Center for Computational and Integrative Biology, MGH, Simches Research Center, Boston, MA 02114, USA
| |
Collapse
|
25
|
Paracoccidioides brasiliensis interferes on dendritic cells maturation by inhibiting PGE2 production. PLoS One 2015; 10:e0120948. [PMID: 25793979 PMCID: PMC4368678 DOI: 10.1371/journal.pone.0120948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/27/2015] [Indexed: 12/03/2022] Open
Abstract
Paracoccidioidomycosis (PCM) is a systemic mycosis, endemic in most Latin American countries, especially in Brazil, whose etiologic agent is the thermodimorphic fungus of the genus Paracoccidioides, comprising cryptic species of Paracoccidioides brasiliensis, S1, PS2, PS3 and Paracoccidioides lutzii. The mechanisms involved in the initial interaction of the fungus with cells of the innate immune response, as dendritic cells (DCs), deserve to be studied. Prostaglandins (PGs) are eicosanoids that play an important role in modulating functions of immune cells including DCs. Here we found that human immature DCs derived from the differentiation of monocytes cultured with GM-CSF and IL-4 release substantial concentrations of PGE2, which, however, were significantly inhibited after challenge with P. brasiliensis. In vitro blocking of pattern recognition receptors (PRRs) by monoclonal antibodies showed the involvement of mannose receptor (MR) in PGE2 inhibition by the fungus. In addition, phenotyping assays showed that after challenge with the fungus, DCs do not change their phenotype of immature cells to mature ones, as well as do not produce IL-12 p70 or adequate concentrations of TNF-α. Assays using exogenous PGE2 confirmed an association between PGE2 inhibition and failure of cells to phenotypically mature in response to P. brasiliensis. We conclude that a P. brasiliensis evasion mechanism exists associated to a dysregulation on DC maturation. These findings may provide novel information for the understanding of the complex interplay between the host and this fungus.
Collapse
|
26
|
Li G, Feng Y, Xu Y, Wu Q, Han Q, Liang X, Yang B, Wang X, Xia X. The anti-infective activity of punicalagin against Salmonella enterica subsp. enterica serovar typhimurium in mice. Food Funct 2015; 6:2357-64. [DOI: 10.1039/c5fo00053j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Punicalagin, a major bioactive component of pomegranate peel, has an anti-infective effect againstS. typhimuriuminfection in mice.
Collapse
Affiliation(s)
- Guanghui Li
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
- College of Food and Biological Engineering
| | - Yuqing Feng
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Yunfeng Xu
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Qian Wu
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Qi'an Han
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Xiujun Liang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Baowei Yang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Xin Wang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Xiaodong Xia
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| |
Collapse
|
27
|
O'Donnell H, McSorley SJ. Salmonella as a model for non-cognate Th1 cell stimulation. Front Immunol 2014; 5:621. [PMID: 25540644 PMCID: PMC4261815 DOI: 10.3389/fimmu.2014.00621] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/21/2014] [Indexed: 01/13/2023] Open
Abstract
Salmonella has been a model pathogen for examining CD4 T cell activation and effector functions for many years due to the strength of the Th1 cell response observed during Salmonella infections, the relative ease of use of Salmonella, the availability of Salmonella-specific T cell reagents, and the well-characterized nature of the model system, the pathogen, and the immune response elicited. Herein, we discuss the use of Salmonella as a model pathogen to explore the complex interaction of T cells with their inflammatory environment. In particular, we address the issue of bystander activation of naïve T cells and non-cognate stimulation of activated and memory T cells. Further, we compare and contrast our current knowledge of these non-cognate responses in CD8 versus CD4 T cells. Finally, we make a case for Salmonella as a particularly appropriate model pathogen in the study of non-cognate CD4 T cell responses based on the strength of the Th1 response during infection, the requirement for CD4 T cells in bacterial clearance, and the well-characterized inflammatory response to conserved molecular patterns induced by Salmonella infection.
Collapse
Affiliation(s)
- Hope O'Donnell
- Department of Anatomy, Physiology and Cell Biology, Center for Comparative Medicine, School of Veterinary Medicine, University of California Davis , Davis, CA , USA ; Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School - Twin Cities , Minneapolis, MN , USA
| | - Stephen J McSorley
- Department of Anatomy, Physiology and Cell Biology, Center for Comparative Medicine, School of Veterinary Medicine, University of California Davis , Davis, CA , USA
| |
Collapse
|
28
|
Grille S, Moreno M, Bascuas T, Marqués JM, Muñoz N, Lens D, Chabalgoity JA. Salmonella enterica serovar Typhimurium immunotherapy for B-cell lymphoma induces broad anti-tumour immunity with therapeutic effect. Immunology 2014; 143:428-37. [PMID: 24834964 DOI: 10.1111/imm.12320] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 11/30/2022] Open
Abstract
Despite the efficacy of current immune-chemotherapy for treatment of B-cell non-Hodgkin lymphoma, a substantial proportion of patients relapse, highlighting the need for new therapeutic modalities. The use of live microorganisms to develop anti-tumoural therapies has evolved since Coley's toxin and is now receiving renewed attention. Salmonella Typhimurium has been shown to be highly effective as an anti-tumour agent in many solid cancer models, but it has not been used in haemato-oncology. Here, we report that intra-tumoural administration of LVR01 (attenuated S. Typhimurium strain with safety profile) elicits local and systemic anti-tumour immunity, resulting in extended survival in a lymphoma model. LVR01 induces intra-tumoural recruitment of neutrophils and activated CD8(+) T cells, as well as increasing the natural killer cell activation status. Furthermore, a systemic specific anti-tumour response with a clear T helper type 1 profile was observed. This approach is an alternative therapeutic strategy for lymphoma patients that could be easily moved into clinical trials.
Collapse
Affiliation(s)
- Sofía Grille
- Laboratory for Vaccine Research, Depto Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Universidad de la República, Montevideo, Uruguay; Departamento Básico de Medicina, Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | | | | | | | | | | | | |
Collapse
|
29
|
The Dynamic Interactions between Salmonella and the Microbiota, within the Challenging Niche of the Gastrointestinal Tract. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:846049. [PMID: 27437481 PMCID: PMC4897363 DOI: 10.1155/2014/846049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/14/2014] [Indexed: 12/25/2022]
Abstract
Understanding how Salmonella species establish successful infections remains a foremost research priority. This gastrointestinal pathogen not only faces the hostile defenses of the host's immune system, but also faces fierce competition from the large and diverse community of microbiota for space and nutrients. Salmonella have solved these challenges ingeniously. To jump-start growth, Salmonella steal hydrogen produced by the gastrointestinal microbiota. Type 3 effector proteins are subsequently secreted by Salmonella to trigger potent inflammatory responses, which generate the alternative terminal electron acceptors tetrathionate and nitrate. Salmonella exclusively utilize these electron acceptors for anaerobic respiration, permitting metabolic access to abundant substrates such as ethanolamine to power growth blooms. Chemotaxis and flagella-mediated motility enable the identification of nutritionally beneficial niches. The resulting growth blooms also promote horizontal gene transfer amongst the resident microbes. Within the gastrointestinal tract there are opportunities for chemical signaling between host cells, the microbiota, and Salmonella. Host produced catecholamines and bacterial autoinducers form components of this chemical dialogue leading to dynamic interactions. Thus, Salmonella have developed remarkable strategies to initially shield against host defenses and to transiently compete against the intestinal microbiota leading to successful infections. However, the immunocompetent host is subsequently able to reestablish control and clear the infection.
Collapse
|
30
|
Kim SP, Park SO, Lee SJ, Nam SH, Friedman M. A Polysaccharide isolated from the liquid culture of Lentinus edodes (Shiitake) mushroom mycelia containing black rice bran protects mice against salmonellosis through upregulation of the Th1 immune reaction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:2384-91. [PMID: 24593132 DOI: 10.1021/jf405223q] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The present study investigated the antibacterial effect of a bioprocessed polysaccharide (BPP) isolated from Lentinus edodes liquid mycelial culture supplemented with black rice bran against murine salmonellosis. BPP was not bactericidal in vitro, it did, however, stimulate uptake of the bacteria into RAW 264.7 murine macrophage cells, as indicated by increased colony-forming unit (CFU) counts of the contents of the lysed macrophages incubated with Salmonella Typhimurium for 30 and 60 min. Two hours postinfection, the bacterial counts drastically increased in the macrophages, but 4 and 8 h postinfection BPP extract-treated cells showed lower bacterial counts than the vehicle (saline phosphate pH 7.4 buffer, PBS)-treated control. BPP elicited altered morphology and markedly elevated inducible nitric oxide (NO) synthase (iNOS) mRNA and protein expression in the infected macrophage cells. BPP also activated leukocytes in S. Typhimurium-infected mice, as determined by spleen lymphocyte proliferation and IFN-γ levels in mice sera. ELISA analysis on cytokine production by Th1 and Th2 immune cells from splenocytes of infected mice showed significant increases in the levels of the following Th1 cytokines: IL-1β, IL-2, IL-6, and IL-12. Histology assays of the livers of mice infected with a sublethal dose (1 × 10(4) CFU) of S. Typhimurium showed that BPP, administered daily through an intraperitoneal (ip) or oral route, protected against necrosis of the liver, a biomarker of in vivo salmonellosis. The lifespan of mice similarly infected with a lethal dose of S. Typhimurium (1 × 10(5) CFU) was significantly extended by ip injection or oral administration of the BPP without side effects. These results suggest that the activity of BPP against bacterial infection in mice occurs mainly through the activation of macrophage-mediated immune response resulting from augmented Th1 immunity. The significance of the results for microbial food safety and human health and further research needs are discussed.
Collapse
Affiliation(s)
- Sung Phil Kim
- Department of Biological Science, Ajou University , Suwon 443-749, Republic of Korea
| | | | | | | | | |
Collapse
|
31
|
Foley SL, Johnson TJ, Ricke SC, Nayak R, Danzeisen J. Salmonella pathogenicity and host adaptation in chicken-associated serovars. Microbiol Mol Biol Rev 2013; 77:582-607. [PMID: 24296573 PMCID: PMC3973385 DOI: 10.1128/mmbr.00015-13] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enteric pathogens such as Salmonella enterica cause significant morbidity and mortality. S. enterica serovars are a diverse group of pathogens that have evolved to survive in a wide range of environments and across multiple hosts. S. enterica serovars such as S. Typhi, S. Dublin, and S. Gallinarum have a restricted host range, in which they are typically associated with one or a few host species, while S. Enteritidis and S. Typhimurium have broad host ranges. This review examines how S. enterica has evolved through adaptation to different host environments, especially as related to the chicken host, and continues to be an important human pathogen. Several factors impact host range, and these include the acquisition of genes via horizontal gene transfer with plasmids, transposons, and phages, which can potentially expand host range, and the loss of genes or their function, which would reduce the range of hosts that the organism can infect. S. Gallinarum, with a limited host range, has a large number of pseudogenes in its genome compared to broader-host-range serovars. S. enterica serovars such as S. Kentucky and S. Heidelberg also often have plasmids that may help them colonize poultry more efficiently. The ability to colonize different hosts also involves interactions with the host's immune system and commensal organisms that are present. Thus, the factors that impact the ability of Salmonella to colonize a particular host species, such as chickens, are complex and multifactorial, involving the host, the pathogen, and extrinsic pressures. It is the interplay of these factors which leads to the differences in host ranges that we observe today.
Collapse
|
32
|
Eri R, Chieppa M. Messages from the Inside. The Dynamic Environment that Favors Intestinal Homeostasis. Front Immunol 2013; 4:323. [PMID: 24130559 PMCID: PMC3793270 DOI: 10.3389/fimmu.2013.00323] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 09/23/2013] [Indexed: 12/15/2022] Open
Abstract
An organism is defined as "an individual living thing capable of responding to stimuli, growing, reproducing, and maintaining homeostasis." Early during evolution multicellular organisms explored the advantages of a symbiotic life. Mammals harbor a complex aggregate of microorganisms (called microbiota) that includes bacteria, fungi, and archaea. Some of these bacteria have already defined beneficial roles for the human host that include the ability to break down nutrients that could not otherwise be digested, preventing the growth of harmful species, as well as the ability to produce vitamins or hormones. It is intuitive that along the evolutionary path several mechanisms favored bacteria that provided advantages to the host which, in return, avoided launching an aggressive immunological response against them. The intestinal immunological response does not ignore the lumenal content, on the contrary, immune surveillance is favored by continuous antigen sampling. Some intestinal epithelial cells (ECs) are crucial during the sampling process, others actively participate in the defense mechanism. In essence the epithelium acts as a traffic light, communicating to the inside world whether conditions are safe or dangerous, and thus influencing immunological response. In this review we will discuss the dynamic factors that act on the intestinal ECs and how they directly or indirectly influence immune cells during states of health and disease.
Collapse
Affiliation(s)
- Rajaraman Eri
- Mucosal Biology, School of Human Life Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Marcello Chieppa
- Laboratory of Experimental Immunopathology, IRCCS “De Bellis,” Castellana Grotte, Bari, Italy
- *Correspondence: Marcello Chieppa, Laboratory of Experimental Immunopathology, IRCCS “De Bellis,” Via Turi 27, Castellana Grotte, Bari 70013, Italy e-mail:
| |
Collapse
|
33
|
Martins RP, Lorenzi V, Arce C, Lucena C, Carvajal A, Garrido JJ. Innate and adaptive immune mechanisms are effectively induced in ileal Peyer's patches of Salmonella typhimurium infected pigs. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:100-104. [PMID: 23644015 DOI: 10.1016/j.dci.2013.04.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/23/2013] [Accepted: 04/24/2013] [Indexed: 06/02/2023]
Abstract
In this report we employed laser-capture microdissection (LCM) coupled to qPCR technology and bioinformatic analysis to characterize, for the first time, the response of Peyer's patches (PP) from orally infected animals to Salmonella typhimurium, in a model of non-typhoidal salmonellosis. Pathogen was highly found in the cytoplasm of phagocytes in PP and differential gene expression analysis indicated an up-regulation of proinflammatory molecules, establishment of a Th1 driven response and triggering of DC and T-cell activity. Furthermore, predictions by bioinformatic analysis pointed to an activation of processes regarding stimulation and maturation of DC, influx of leukocytes in tissue and T lymphocytes priming and differentiation. In short, the approach used in this study proved to be a promising strategy to explore infectious processes. Indeed, it revealed an effective induction of innate and adaptive immune mechanisms in swine PP which appear to be distinct from those observed in mesenteric lymph nodes and closely related to response of gut mucosa.
Collapse
Affiliation(s)
- Rodrigo Prado Martins
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Campus de Rabanales, Edificio Gregor Mendel C5, 14071 Córdoba, Spain
| | | | | | | | | | | |
Collapse
|
34
|
In vivo targeting of activated leukocytes by a β2-integrin binding peptide. Mol Diagn Ther 2013; 18:39-44. [PMID: 23982749 DOI: 10.1007/s40291-013-0052-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND In immunopathological conditions, clinical diagnosis is commonly made on the basis of patient symptoms, measurement of blood leukocyte levels or proinflammatory biomarkers, non-specific radiological findings and, regarding infection, microbiological analysis. Nevertheless, frequently the exact spatial location of inflammation or even infection cannot be reliably identified, despite the use of up-to-date clinical, laboratory and imaging techniques. For this reason, new tools are warranted for use in advanced diagnosis and therapy targeting in patients. OBJECTIVE The peptide CPCFLLGCC (LLG), known to bind activated β2-integrins in vitro, was fused with green fluorescent protein (GFP) to test the ability of LLG fusions to target and bind activated leukocytes in vivo. METHODS A murine skin scratch inflammation model was chosen for the convenient non-surgical detection of GFP. RESULTS The murine skin lesion inflammation model demonstrated in vivo targeting of LLG-GFP to sites of inflammation. Targeting by LLG-GFP fusion construct depends on the ability of the LLG-moiety to bind activated leukocytes. Control construct unable to bind β2-integrins appeared biologically inert. CONCLUSION The data support the possibility of using this fluorescently labeled peptide as a tool for both the detection of and targeting to inflammatory sites characterized by robust leukocyte activation.
Collapse
|
35
|
Tardif V, Riquelme SA, Remy S, Carreño LJ, Cortés CM, Simon T, Hill M, Louvet C, Riedel CA, Blancou P, Bach JM, Chauveau C, Bueno SM, Anegon I, Kalergis AM. Carbon monoxide decreases endosome-lysosome fusion and inhibits soluble antigen presentation by dendritic cells to T cells. Eur J Immunol 2013; 43:2832-44. [PMID: 23852701 DOI: 10.1002/eji.201343600] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/27/2013] [Accepted: 07/11/2013] [Indexed: 11/06/2022]
Abstract
Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. We have previously shown that either induction of HO-1 or treatment with exogenous CO inhibits LPS-induced maturation of dendritic cells (DCs) and protects in vivo and in vitro antigen-specific inflammation. Here, we evaluated the capacity of HO-1 and CO to regulate antigen presentation on MHC class I and MHC class II molecules by LPS-treated DCs. We observed that HO-1 and CO treatment significantly inhibited the capacity of DCs to present soluble antigens to T cells. Inhibition was restricted to soluble OVA protein, as no inhibition was observed for antigenic OVA-derived peptides, bead-bound OVA protein, or OVA as an endogenous antigen. Inhibition of soluble antigen presentation was not due to reduced antigen uptake by DCs, as endocytosis remained functional after HO-1 induction and CO treatment. On the contrary, CO significantly reduced the efficiency of fusion between late endosomes and lysosomes and not by phagosomes and lysosomes. These data suggest that HO-1 and CO can inhibit the ability of LPS-treated DCs to present exogenous soluble antigens to naïve T cells by blocking antigen trafficking at the level of late endosome-lysosome fusion.
Collapse
Affiliation(s)
- Virginie Tardif
- INSERM, UMR 1064, Nantes, France; CHU Nantes, ITUN, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fabrik I, Härtlova A, Rehulka P, Stulik J. Serving the new masters - dendritic cells as hosts for stealth intracellular bacteria. Cell Microbiol 2013; 15:1473-83. [PMID: 23795643 DOI: 10.1111/cmi.12160] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/14/2013] [Accepted: 05/23/2013] [Indexed: 02/01/2023]
Abstract
Dendritic cells (DCs) serve as the primers of adaptive immunity, which is indispensable for the control of the majority of infections. Interestingly, some pathogenic intracellular bacteria can subvert DC function and gain the advantage of an ineffective host immune reaction. This scenario appears to be the case particularly with so-called stealth pathogens, which are the causative agents of several under-diagnosed chronic diseases. However, there is no consensus how less explored stealth bacteria like Coxiella, Brucella and Francisella cross-talk with DCs. Therefore, the aim of this review was to explore the issue and to summarize the current knowledge regarding the interaction of above mentioned pathogens with DCs as crucial hosts from an infection strategy view. Evidence indicates that infected DCs are not sufficiently activated, do not undergo maturation and do not produce expected proinflammatory cytokines. In some cases, the infected DCs even display immunosuppressive behaviour that may be directly linked to the induction of tolerogenicity favouring pathogen survival and persistence.
Collapse
Affiliation(s)
- Ivo Fabrik
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic.
| | | | | | | |
Collapse
|
37
|
Dunne KA, Allam A, McIntosh A, Houston SA, Cerovic V, Goodyear CS, Roe AJ, Beatson SA, Milling SW, Walker D, Wall DM. Increased S-nitrosylation and proteasomal degradation of caspase-3 during infection contribute to the persistence of adherent invasive Escherichia coli (AIEC) in immune cells. PLoS One 2013; 8:e68386. [PMID: 23861899 PMCID: PMC3701656 DOI: 10.1371/journal.pone.0068386] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 06/04/2013] [Indexed: 01/12/2023] Open
Abstract
Adherent invasive Escherichia coli (AIEC) have been implicated as a causative agent of Crohn’s disease (CD) due to their isolation from the intestines of CD sufferers and their ability to persist in macrophages inducing granulomas. The rapid intracellular multiplication of AIEC sets it apart from other enteric pathogens such as Salmonella Typhimurium which after limited replication induce programmed cell death (PCD). Understanding the response of infected cells to the increased AIEC bacterial load and associated metabolic stress may offer insights into AIEC pathogenesis and its association with CD. Here we show that AIEC persistence within macrophages and dendritic cells is facilitated by increased proteasomal degradation of caspase-3. In addition S-nitrosylation of pro- and active forms of caspase-3, which can inhibit the enzymes activity, is increased in AIEC infected macrophages. This S-nitrosylated caspase-3 was seen to accumulate upon inhibition of the proteasome indicating an additional role for S-nitrosylation in inducing caspase-3 degradation in a manner independent of ubiquitination. In addition to the autophagic genetic defects that are linked to CD, this delay in apoptosis mediated in AIEC infected cells through increased degradation of caspase-3, may be an essential factor in its prolonged persistence in CD patients.
Collapse
Affiliation(s)
- Karl A. Dunne
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Amr Allam
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Anne McIntosh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Stephanie A. Houston
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Vuk Cerovic
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Carl S. Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andrew J. Roe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Scott A. Beatson
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, University of Queensland, St. Lucia, Queensland, Australia
| | - Simon W. Milling
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Daniel Walker
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Daniel M. Wall
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Jackson NP, Kang YH, Lapaque N, Janssen H, Trowsdale J, Kelly AP. Salmonella polarises peptide-MHC-II presentation towards an unconventional Type B CD4+ T-cell response. Eur J Immunol 2013; 43:897-906. [PMID: 23319341 PMCID: PMC3816330 DOI: 10.1002/eji.201242983] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 12/03/2012] [Accepted: 01/08/2013] [Indexed: 11/07/2022]
Abstract
Distinct peptide-MHC-II complexes, recognised by Type A and B CD4(+) T-cell subsets, are generated when antigen is loaded in different intracellular compartments. Conventional Type A T cells recognize their peptide epitope regardless of the route of processing, whereas unconventional Type B T cells only recognise exogenously supplied peptide. Type B T cells are implicated in autoimmune conditions and may break tolerance by escaping negative selection. Here we show that Salmonella differentially influences presentation of antigen to Type A and B T cells. Infection of bone marrow-derived dendritic cells (BMDCs) with Salmonella enterica serovar Typhimurium (S. Typhimurium) reduced presentation of antigen to Type A T cells but enhanced presentation of exogenous peptide to Type B T cells. Exposure to S. Typhimurium was sufficient to enhance Type B T-cell activation. Salmonella Typhimurium infection reduced surface expression of MHC-II, by an invariant chain-independent trafficking mechanism, resulting in accumulation of MHC-II in multi-vesicular bodies. Reduced MHC-II surface expression in S. Typhimurium-infected BMDCs correlated with reduced antigen presentation to Type A T cells. Salmonella infection is implicated in reactive arthritis. Therefore, polarisation of antigen presentation towards a Type B response by Salmonella may be a predisposing factor in autoimmune conditions such as reactive arthritis.
Collapse
Affiliation(s)
- Nicola P Jackson
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|