1
|
Sharma N, Das A, Nair AV, Sethi P, Negi VD, Chakravortty D, Marathe SA. CRISPR-Cas system positively regulates virulence of Salmonella enterica serovar Typhimurium. Gut Pathog 2024; 16:63. [PMID: 39462402 PMCID: PMC11514906 DOI: 10.1186/s13099-024-00653-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/06/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Salmonella, a foodborne pathogen, possesses a type I-E clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated (Cas) system. We investigated the system's role in regulating Salmonella virulence by deleting the CRISPR arrays and Cas operon. RESULTS Our study demonstrates invasion and proliferation defects of CRISPR-Cas knockout strains in intestinal epithelial cells and macrophages owing to the repression of invasion and virulence genes. However, proliferation defects were not observed in the Gp91phox-/- macrophages, suggesting the system's role in the pathogens' antioxidant defense. We deduced that the CRISPR-Cas system positively regulates H2O2 importer (OmpW), catalase (katG), peroxidase (ahpC), and superoxide dismutase (soda and sodCI), thereby protecting the cells from oxidative radicals. The knockout strains were attenuated in in-vivo infection models (Caenorhabditis elegans and BALB/c mice) due to hypersensitivity against antimicrobial peptides, complement proteins, and oxidative stress. The attenuation in virulence was attributed to the suppression of LPS modifying (pmr) genes, antioxidant genes, master regulators, and effectors of the SPI-1 (invasion) and SPI-2 (proliferation) islands in knockout strains. The regulation could be attributed to the partial complementarity of the CRISPR spacers with these genes. CONCLUSIONS Overall, our study extends our understanding of the role of the CRISPR-Cas system in Salmonella pathogenesis and its virulence determinants.
Collapse
Affiliation(s)
- Nandita Sharma
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, 333031, India
| | - Ankita Das
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, 333031, India
| | - Abhilash Vijay Nair
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Palash Sethi
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, 333031, India
| | - Vidya Devi Negi
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, 140306, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Sandhya Amol Marathe
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
2
|
Singh S, Koo OK. A Comprehensive Review Exploring the Protective Role of Specific Commensal Gut Bacteria against Salmonella. Pathogens 2024; 13:642. [PMID: 39204243 PMCID: PMC11356920 DOI: 10.3390/pathogens13080642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Gut microbiota is a diverse community of microorganisms that constantly work to protect the gut against pathogens. Salmonella stands out as a notorious foodborne pathogen that interacts with gut microbes, causing an imbalance in the overall composition of microbiota and leading to dysbiosis. This review focuses on the interactions between Salmonella and the key commensal bacteria such as E. coli, Lactobacillus, Clostridium, Akkermansia, and Bacteroides. The review highlights the role of these gut bacteria and their synergy in combating Salmonella through several mechanistic interactions. These include the production of siderophores, which compete with Salmonella for essential iron; the synthesis of short-chain fatty acids (SCFAs), which exert antimicrobial effects and modulate the gut environment; the secretion of bacteriocins, which directly inhibit Salmonella growth; and the modulation of cytokine responses, which influences the host's immune reaction to infection. While much research has explored Salmonella, this review aims to better understand how specific gut bacteria engage with the pathogen, revealing distinct defense mechanisms tailored to each species and how their synergy may lead to enhanced protection against Salmonella. Furthermore, the combination of these commensal bacteria could offer promising avenues for bacteria-mediated therapy during Salmonella-induced gut infections in the future.
Collapse
Affiliation(s)
| | - Ok Kyung Koo
- Department of Food Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
3
|
Lynch CA, Acosta SA, Anderson DM, Rogers GE, Wilson-Rawls J, Rawls A. The Transcription Factor Mohawk Facilitates Skeletal Muscle Repair via Modulation of the Inflammatory Environment. Int J Mol Sci 2024; 25:5019. [PMID: 38732238 PMCID: PMC11084535 DOI: 10.3390/ijms25095019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
Efficient repair of skeletal muscle relies upon the precise coordination of cells between the satellite cell niche and innate immune cells that are recruited to the site of injury. The expression of pro-inflammatory cytokines and chemokines such as TNFα, IFNγ, CXCL1, and CCL2, by muscle and tissue resident immune cells recruits neutrophils and M1 macrophages to the injury and activates satellite cells. These signal cascades lead to highly integrated temporal and spatial control of muscle repair. Despite the therapeutic potential of these factors for improving tissue regeneration after traumatic and chronic injuries, their transcriptional regulation is not well understood. The transcription factor Mohawk (Mkx) functions as a repressor of myogenic differentiation and regulates fiber type specification. Embryonically, Mkx is expressed in all progenitor cells of the musculoskeletal system and is expressed in human and mouse myeloid lineage cells. An analysis of mice deficient for Mkx revealed a delay in postnatal muscle repair characterized by impaired clearance of necrotic fibers and smaller newly regenerated fibers. Further, there was a delay in the expression of inflammatory signals such as Ccl2, Ifnγ, and Tgfß. This was coupled with impaired recruitment of pro-inflammatory macrophages to the site of muscle damage. These studies demonstrate that Mkx plays a critical role in adult skeletal muscle repair that is mediated through the initial activation of the inflammatory response.
Collapse
Affiliation(s)
- Cherie Alissa Lynch
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Sofia A. Acosta
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Douglas M. Anderson
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Gavin E. Rogers
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ 85287, USA
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85287, USA; (C.A.L.); (S.A.A.); (D.M.A.); (G.E.R.); (J.W.-R.)
| |
Collapse
|
4
|
Porwollik S, Chu W, Desai PT, McClelland M. A genome-wide collection of barcoded single-gene deletion mutants in Salmonella enterica serovar Typhimurium. PLoS One 2024; 19:e0298419. [PMID: 38452024 PMCID: PMC10919679 DOI: 10.1371/journal.pone.0298419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024] Open
Abstract
Genetic screening of pools of mutants can reveal genetic determinants involved in complex biological interactions, processes, and systems. We previously constructed two single-gene deletion resources for Salmonella enterica serovar Typhimurium 14028s in which kanamycin (KanR) and chloramphenicol (CamR) cassettes were used to replace non-essential genes. We have now used lambda-red recombination to convert the antibiotic cassettes in these resources into a tetracycline-resistant (TetR) version where each mutant contains a different 21-base barcode flanked by Illumina Read1 and Read2 primer sequences. A motility assay of a pool of the entire library, followed by a single-tube processing of the bacterial pellet, PCR, and sequencing, was used to verify the performance of the barcoded TetR collection. The new resource is useful for experiments with defined subsets of barcoded mutant strains where biological bottlenecks preclude high numbers of founder bacteria, such as in animal infections. The TetR version of the library will also facilitate the construction of triple mutants by transduction. The resource of 6197 mutants covering 3490 genes is deposited at Biological and Emerging Infections Resources (beiresources.org).
Collapse
Affiliation(s)
- Steffen Porwollik
- Dept. of Microbiology and Molecular Genetics, University of California, Irvine, Irvina, CA, United States of America
| | - Weiping Chu
- Dept. of Microbiology and Molecular Genetics, University of California, Irvine, Irvina, CA, United States of America
| | - Prerak T. Desai
- GSK Computational Biology, Upper Providence, PA, United States of America
| | - Michael McClelland
- Dept. of Microbiology and Molecular Genetics, University of California, Irvine, Irvina, CA, United States of America
| |
Collapse
|
5
|
Wongkuna S, Ambat A, Ghimire S, Mattiello SP, Maji A, Kumar R, Antony L, Chankhamhaengdecha S, Janvilisri T, Nelson E, Doerner KC, More S, Behr M, Scaria J. Identification of a microbial sub-community from the feral chicken gut that reduces Salmonella colonization and improves gut health in a gnotobiotic chicken model. Microbiol Spectr 2024; 12:e0162123. [PMID: 38315031 PMCID: PMC10913435 DOI: 10.1128/spectrum.01621-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 12/16/2023] [Indexed: 02/07/2024] Open
Abstract
A complex microbial community in the gut may prevent the colonization of enteric pathogens such as Salmonella. Some individual or a combination of species in the gut may confer colonization resistance against Salmonella. To gain a better understanding of the colonization resistance against Salmonella enterica, we isolated a library of 1,300 bacterial strains from feral chicken gut microbiota which represented a total of 51 species. Using a co-culture assay, we screened the representative species from this library and identified 30 species that inhibited Salmonella enterica subspecies enterica serovar Typhimurium in vitro. To improve the Salmonella inhibition capacity, from a pool of fast-growing species, we formulated 66 bacterial blends, each of which composed of 10 species. Bacterial blends were more efficient in inhibiting Salmonella as compared to individual species. The blend that showed maximum inhibition (Mix10) also inhibited other serotypes of Salmonella frequently found in poultry. The in vivo effect of Mix10 was examined in a gnotobiotic and conventional chicken model. The Mix10 consortium significantly reduced Salmonella load at day 2 post-infection in gnotobiotic chicken model and decreased intestinal tissue damage and inflammation in both models. Cell-free supernatant of Mix10 did not show Salmonella inhibition, indicating that Mix10 inhibits Salmonella through either nutritional competition, competitive exclusion, or through reinforcement of host immunity. Out of 10 species, 3 species in Mix10 did not colonize, while 3 species constituted more than 70% of the community. Two of these species were previously uncultured bacteria. Our approach could be used as a high-throughput screening system to identify additional bacterial sub-communities that confer colonization resistance against enteric pathogens and its effect on the host.IMPORTANCESalmonella colonization in chicken and human infections originating from Salmonella-contaminated poultry is a significant problem. Poultry has been identified as the most common food linked to enteric pathogen outbreaks in the United States. Since multi-drug-resistant Salmonella often colonize chicken and cause human infections, methods to control Salmonella colonization in poultry are needed. The method we describe here could form the basis of developing gut microbiota-derived bacterial blends as a microbial ecosystem therapeutic against Salmonella.
Collapse
Affiliation(s)
- Supapit Wongkuna
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Achuthan Ambat
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sudeep Ghimire
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Samara Paula Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Abhijit Maji
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Roshan Kumar
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Linto Antony
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | | | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Eric Nelson
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Kinchel C. Doerner
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Sunil More
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Melissa Behr
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, South Dakota, USA
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
6
|
Lee JS, Kim C. Role of CARD9 in Cell- and Organ-Specific Immune Responses in Various Infections. Int J Mol Sci 2024; 25:2598. [PMID: 38473845 DOI: 10.3390/ijms25052598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
The caspase recruitment domain-containing protein 9 (CARD9) is an intracellular adaptor protein that is abundantly expressed in cells of the myeloid lineage, such as neutrophils, macrophages, and dendritic cells. CARD9 plays a critical role in host immunity against infections caused by fungi, bacteria, and viruses. A CARD9 deficiency impairs the production of inflammatory cytokines and chemokines as well as migration and infiltration, thereby increasing susceptibility to infections. However, CARD9 signaling varies depending on the pathogen causing the infection. Furthermore, different studies have reported altered CARD9-mediated signaling even with the same pathogen. Therefore, this review focuses on and elucidates the current literature on varied CARD9 signaling in response to various infectious stimuli in humans and experimental mice models.
Collapse
Affiliation(s)
- Ji Seok Lee
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, Inha University School of Medicine, Incheon 22212, Republic of Korea
- BK21, Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, Inha University School of Medicine, Incheon 22212, Republic of Korea
- BK21, Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
7
|
Milby-Blackledge A, Farnell Y, Zhao D, Berghman L, Laino C, Muller M, Byrd JA, Farnell M. Serum cytokine profile of neonatal broiler chickens infected with Salmonella Typhimurium. Front Physiol 2024; 15:1359722. [PMID: 38465263 PMCID: PMC10920336 DOI: 10.3389/fphys.2024.1359722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024] Open
Abstract
The avian immune system responds to Salmonella infection by expressing cytokines and chemokines. We hypothesized that the immune status of Salmonella Typhimurium (ST) challenged neonatal broilers would differ from the uninfected treatment. The objective of this experiment was to evaluate 12 cytokines. Day of hatch male chicks were randomly allocated into a control or ST challenged group. At day three of age, sterile diluent or 5.0 × 108 CFU of ST was given orally to each chick. Blood was obtained 24 h post challenge and serum separated for later analysis (n = 30 chicks/treatment). Significant (p ≤ 0.05) increases in pro-inflammatory cytokines-interleukin-6 (IL-6), IL-16, and IL-21; anti-inflammatory cytokines- IL-10; chemokines-regulated on activation, normal T cell expressed and secreted (RANTES), macrophage inflammatory protein-1β (MIP-1β), and MIP-3α; colony stimulating factors-macrophage colony-stimulating factor (M-CSF); and growth factors-vascular endothelial growth factor (VEGF) were observed in the serum of the challenged chicks when compared to the control. No significant differences were observed in IL-2, interferon gamma (IFNγ), and IFNα. These data indicate the detection of mucosal immune responses in broiler chickens following ST infection. The heightened levels of pro-inflammatory cytokines, chemokines, and colony stimulating factors align with known inflammatory mechanisms, like the influx of immune cells. However, the elevation of IL-10 was unexpected, due to its immunoregulatory properties. Notably, the rise in VEGF levels is compelling, as it suggests the possibility of tissue repair and angiogenesis in ST infected birds.
Collapse
Affiliation(s)
| | - Yuhua Farnell
- Texas A&M AgriLife Research, Department of Poultry Science, College Station, TX, United States
| | - Dan Zhao
- Texas A&M AgriLife Research, Department of Poultry Science, College Station, TX, United States
| | - Luc Berghman
- Texas A&M AgriLife Research, Department of Poultry Science, College Station, TX, United States
| | - Craig Laino
- Millipore Sigma, Saint Louis, MO, United States
| | | | - J. Allen Byrd
- United States Department of Agriculture, Southern Plains Agricultural Research Service, College Station, TX, United States
| | - Morgan Farnell
- Texas A&M AgriLife Research, Department of Poultry Science, College Station, TX, United States
| |
Collapse
|
8
|
Valini GADC, Méthot S, Pomar C, Hauschild L, Remus A. Size matters: lower body weight pigs have a different response to immune challenge and amino acids supplementation above the estimated requirement compared to heavy pigs. J Anim Sci 2024; 102:skae255. [PMID: 39207124 PMCID: PMC11439151 DOI: 10.1093/jas/skae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
The immune response varies between pigs, as not all pigs have the same response to a stressor. This variation may exist between individuals due to body weight (BW) or body composition, which may impact the capacity for coping with an immune challenge (IC). Tryptophan (Trp), threonine (Thr), and methionine (Met) requirements might also play a considerable part in supporting immune system activation while reducing variation between pigs; however, the latter has yet to be reported. This exploratory study investigated the effect of initial BW (light vs. heavy-weight) and supplementation of Trp, Thr, and Met above National Research Council (NRC) requirements on feeding behavior and the coping capacity of growing pigs under an IC. Eighty gilts were categorized into 2 groups according to BW: light-weight (LW, 22.5 kg) and heavy-weight pigs (HW, 28.5 kg). Both BW groups were group-housed for a 28-d trial in a good or poor sanitary condition (SC). Pigs within a poor SC were orally inoculated with 2 × 109 colony units of Salmonella Typhimurium, and fresh manure from a pig farm was spread on the floor. Pigs within good SC were not inoculated, nor was manure spread. Two diets were provided within each SC: control (CN) or supplemented (AA+) with Trp, Thr, and Met at 120% of NRC recommended levels. A principal component analysis was performed in R, and a feeding behavior index was calculated in SAS. Results showed that LW and HW pigs were clustered separately on day 0, where LW pigs had a positive correlation with body lipid percentage (r = 0.83), and HW pigs had a positive correlation with body protein percentage (r = 0.75). After the IC, the cluster configuration changed, with diets influencing LW more than HW pigs within poor SC. On day 14, LW fed AA + diet in poor SC was clustered separately from LW pigs fed CN diet, whereas LW fed AA + and CN diets in good SC were clustered together. For feeding behavior, in both analyzed periods (period 1: days 7 to 14; period 2: days 21 to 28), LW had lower total feed intake and shorter meals than HW pigs (P < 0.10), independent of the SC. Furthermore, LW pigs fed AA + diet had a more regular feed intake pattern than those fed CN diet, while a more irregular pattern was observed for HW pigs fed AA + diet than CN diet at period 2. These findings suggest that supplementing Trp, Thr, and Met above requirements may be a nutritional strategy for LW pigs under IC by improving feed intake regularity and reducing the probability of being susceptible to IC.
Collapse
Affiliation(s)
- Graziela Alves da Cunha Valini
- Department of Animal Science, School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal, SP, Brazil
- Sherbrooke Research and Development Centre – Agriculture and Agri-Food Canada, Sherbrooke, Québec, Canada J1M 0C8
| | - Steve Méthot
- Sherbrooke Research and Development Centre – Agriculture and Agri-Food Canada, Sherbrooke, Québec, Canada J1M 0C8
| | - Candido Pomar
- Sherbrooke Research and Development Centre – Agriculture and Agri-Food Canada, Sherbrooke, Québec, Canada J1M 0C8
| | - Luciano Hauschild
- Department of Animal Science, School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal, SP, Brazil
| | - Aline Remus
- Sherbrooke Research and Development Centre – Agriculture and Agri-Food Canada, Sherbrooke, Québec, Canada J1M 0C8
| |
Collapse
|
9
|
Teixeira SC, Teixeira TL, Tavares PCB, Alves RN, da Silva AA, Borges BC, Martins FA, Dos Santos MA, de Castilhos P, E Silva Brígido RT, Notário AFO, Silveira ACA, da Silva CV. Subversion strategies of lysosomal killing by intracellular pathogens. Microbiol Res 2023; 277:127503. [PMID: 37748260 DOI: 10.1016/j.micres.2023.127503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/27/2023]
Abstract
Many pathogenic organisms need to reach either an intracellular compartment or the cytoplasm of a target cell for their survival, replication or immune system evasion. Intracellular pathogens frequently penetrate into the cell through the endocytic and phagocytic pathways (clathrin-mediated endocytosis, phagocytosis and macropinocytosis) that culminates in fusion with lysosomes. However, several mechanisms are triggered by pathogenic microorganisms - protozoan, bacteria, virus and fungus - to avoid destruction by lysosome fusion, such as rupture of the phagosome and thereby release into the cytoplasm, avoidance of autophagy, delaying in both phagolysosome biogenesis and phagosomal maturation and survival/replication inside the phagolysosome. Here we reviewed the main data dealing with phagosome maturation and evasion from lysosomal killing by different bacteria, protozoa, fungi and virus.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Aline Alves da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Flávia Alves Martins
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marlus Alves Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Patrícia de Castilhos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
10
|
Shaji S, Selvaraj RK, Shanmugasundaram R. Salmonella Infection in Poultry: A Review on the Pathogen and Control Strategies. Microorganisms 2023; 11:2814. [PMID: 38004824 PMCID: PMC10672927 DOI: 10.3390/microorganisms11112814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/08/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Salmonella is the leading cause of food-borne zoonotic disease worldwide. Non-typhoidal Salmonella serotypes are the primary etiological agents associated with salmonellosis in poultry. Contaminated poultry eggs and meat products are the major sources of human Salmonella infection. Horizontal and vertical transmission are the primary routes of infection in chickens. The principal virulence genes linked to Salmonella pathogenesis in poultry are located in Salmonella pathogenicity islands 1 and 2 (SPI-1 and SPI-2). Cell-mediated and humoral immune responses are involved in the defense against Salmonella invasion in poultry. Vaccination of chickens and supplementation of feed additives like prebiotics, probiotics, postbiotics, synbiotics, and bacteriophages are currently being used to mitigate the Salmonella load in poultry. Despite the existence of various control measures, there is still a need for a broad, safe, and well-defined strategy that can confer long-term protection from Salmonella in poultry flocks. This review examines the current knowledge on the etiology, transmission, cell wall structure, nomenclature, pathogenesis, immune response, and efficacy of preventative approaches to Salmonella.
Collapse
Affiliation(s)
- Syamily Shaji
- Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA; (S.S.); (R.K.S.)
| | - Ramesh K. Selvaraj
- Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA; (S.S.); (R.K.S.)
| | - Revathi Shanmugasundaram
- Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA
| |
Collapse
|
11
|
Su Y, Sun T, Gao J, Zhang C, Liu X, Bi C, Wang J, Shan A. Anti-Proteolytic Peptide R7I Protects the Intestinal Barrier and Alleviates Fatty Acid Malabsorption in Salmonella typhimurium-Infected Mice. Int J Mol Sci 2023; 24:16409. [PMID: 38003599 PMCID: PMC10670956 DOI: 10.3390/ijms242216409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
With a wide range of hosts, environmental adaptation, and antibiotic resistance, Salmonella typhimurium is one of the most common causes of food poisoning in the world. Infection with Salmonella typhimurium not only results in intestinal inflammation but also damages the intestinal barrier and interferes with the host's ability to absorb nutrients. It is imperative to find alternatives to antibiotics for eradicating bacteria, reducing intestinal damage, and reestablishing nutrient absorption, especially given that antibiotics are currently prohibited. This research aims to understand the protective role of anti-proteolytic peptide R7I on the gut in the setting of Salmonella typhimurium infection and its impact on nutritional absorption, maybe offering an alternative to antibiotics for bacterial killing. The findings demonstrated that R7I reduced the production of inflammatory factors, including IL-6, TNF-α, and L-1β in the jejunum and decreased the expression of genes like TLR4 and NF-κB in the jejunum (p < 0.05). R7I enhanced antioxidant capacity and preserved the antioxidant/pro-oxidant balance in the jejunum (p < 0.05). R7I also normalized intestinal shape and restored tight junction protein expression. Fatty acid binding protein 2 (FABP2) and fatty acid transport protein 4 (FATP4) expression in the jejunum was restored by R7I. In addition, serum-free fatty acids and lipid metabolites were significantly higher in the R7I group than in the control group (p < 0.05). Overall, the anti-enzyme peptide R7I maintained the healthy state of the intestine and alleviated the abnormal fatty acid absorption caused by bacterial infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jiajun Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Y.S.); (T.S.); (J.G.); (C.Z.); (X.L.); (C.B.)
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (Y.S.); (T.S.); (J.G.); (C.Z.); (X.L.); (C.B.)
| |
Collapse
|
12
|
El-Shiekh RA, Elshimy R. Therapeutic effects of Stemmoside C against Salmonella enterica serotype typhimurium infected BALB/c mice. Steroids 2023; 199:109296. [PMID: 37591445 DOI: 10.1016/j.steroids.2023.109296] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Salmonella is a Gram-negative bacterium that causes gastrointestinal diseases in 20 to 40 million people globally. Stemmoside C is a steroidal glycoside isolated from Argel, although its antibacterial and antibiofilm properties have not been studied. The antibacterial activity of Stemmoside C against Salmonella enterica was revealed, where MIC of the compound was 16 μg/mL (0.15 µM). Biofilm-associated Stemmoside C treatment destroyed S. typhi cells and reduced viable S. typhi numbers below detectable levels. When compared to Stemmoside C or Ciprofluxacin-treated mice, infected BALB/c mice had a greater death rate and a larger bacterial blood burden. The protective effects of orally administered Stemmoside C at dose of 25 and 50 mg/kg b.wt. against bacterial infection was associated with reduction in the levels of inflammatory cytokines (IFN-γ, Il-1β, IL-2, IL-6, MPO, and TNF-α) and elevation of anti-inflammatory cytokine (IL-10 and IL-12) in serum. Where, Stemmoside C at dose of 50 mg/kg b.wt. regulated the levels almost as normal control group and demonstrated apparently normal intestinal sections. It also resulted in a decrease in the number of viable S. typhi retrieved from feces. Stemmoside C is a promising drug for the treatment or prevention of S. typhimurium infection.
Collapse
Affiliation(s)
- Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr el Aini st., Cairo 11562, Egypt.
| | - Rana Elshimy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt; Department of Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt.
| |
Collapse
|
13
|
Chen H, Cao Z, Liu M, Diamond MS, Jin X. The impact of helminth-induced immunity on infection with bacteria or viruses. Vet Res 2023; 54:87. [PMID: 37789420 PMCID: PMC10548622 DOI: 10.1186/s13567-023-01216-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/21/2023] [Indexed: 10/05/2023] Open
Abstract
Different human and animal pathogens trigger distinct immune responses in their hosts. The infection of bacteria or viruses can trigger type I pro-inflammatory immune responses (e.g., IFN-γ, TNF-α, TH1 cells), whereas infection by helminths typically elicits a type II host resistance and tolerizing immune response (e.g., IL-4, IL-5, IL-13, TH2 cells). In some respects, the type I and II immune responses induced by these different classes of pathogens are antagonistic. Indeed, recent studies indicate that infection by helminths differentially shapes the response and outcome of subsequent infection by viruses and bacteria. In this review, we summarize the current knowledge on how helminth infections influence concurrent or subsequent microbial infections and also discuss the implications for helminth-mediated immunity on the outcome of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Hong Chen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengguo Cao
- State Key Laboratory of Virology, Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
14
|
Pokorzynski ND, Groisman EA. How Bacterial Pathogens Coordinate Appetite with Virulence. Microbiol Mol Biol Rev 2023; 87:e0019822. [PMID: 37358444 PMCID: PMC10521370 DOI: 10.1128/mmbr.00198-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Cells adjust growth and metabolism to nutrient availability. Having access to a variety of carbon sources during infection of their animal hosts, facultative intracellular pathogens must efficiently prioritize carbon utilization. Here, we discuss how carbon source controls bacterial virulence, with an emphasis on Salmonella enterica serovar Typhimurium, which causes gastroenteritis in immunocompetent humans and a typhoid-like disease in mice, and propose that virulence factors can regulate carbon source prioritization by modifying cellular physiology. On the one hand, bacterial regulators of carbon metabolism control virulence programs, indicating that pathogenic traits appear in response to carbon source availability. On the other hand, signals controlling virulence regulators may impact carbon source utilization, suggesting that stimuli that bacterial pathogens experience within the host can directly impinge on carbon source prioritization. In addition, pathogen-triggered intestinal inflammation can disrupt the gut microbiota and thus the availability of carbon sources. By coordinating virulence factors with carbon utilization determinants, pathogens adopt metabolic pathways that may not be the most energy efficient because such pathways promote resistance to antimicrobial agents and also because host-imposed deprivation of specific nutrients may hinder the operation of certain pathways. We propose that metabolic prioritization by bacteria underlies the pathogenic outcome of an infection.
Collapse
Affiliation(s)
- Nick D. Pokorzynski
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Eduardo A. Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Microbial Sciences Institute, West Haven, Connecticut, USA
| |
Collapse
|
15
|
Wu S, Zhang Q, Cong G, Xiao Y, Shen Y, Zhang S, Zhao W, Shi S. Probiotic Escherichia coli Nissle 1917 protect chicks from damage caused by Salmonella enterica serovar Enteritidis colonization. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:450-460. [PMID: 37649679 PMCID: PMC10463197 DOI: 10.1016/j.aninu.2023.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/14/2023] [Accepted: 06/02/2023] [Indexed: 09/01/2023]
Abstract
As a foodborne pathogen of global importance, Salmonella enterica serovar Enteritidis (S. Enteritidis) is a threat to public health that is mainly spread by poultry products. Intestinal Enterobacteriaceae can inhibit the colonization of S. Enteritidis and are regarded as a potential antibiotic substitute. We investigated, in chicks, the anti-S. Enteritidis effects of Escherichia coli (E. coli) Nissle 1917, the most well-known probiotic member of Enterobacteriaceae. Eighty 1-d-old healthy female AA broilers were randomly divided into 4 groups, with 20 in each group, namely the negative control (group P), the E. coli Nissle 1917-treated group (group N), the S. Enteritidis-infected group (group S) and the E. coli Nissle 1917-treated and S. Enteritidis-infected group (group NS). From d 5 to 7, chicks in groups N and NS were orally gavaged once a day with E. coli Nissle 1917 and in groups P and S were administered the same volume of sterile PBS. At d 8, the chicks in groups S and NS were orally gavaged with S. Enteritidis and in groups P and N were administered the same volume of sterile PBS. Sampling was conducted 24 h after challenge. Results showed that gavage of E. coli Nissle 1917 reduced the spleen index, Salmonella loads, and inflammation (P < 0.05). It improved intestinal morphology and intestinal barrier function (P < 0.05). S. Enteritidis infection significantly reduced mRNA expression of angiotensin-converting enzyme 2 (ACE2) and solute carrier family 6-member 19 (SLC6A19) in the cecum and the content of Gly, Ser, Gln, and Trp in the serum (P < 0.05). Pretreatment with E. coli Nissle 1917 yielded mRNA expression of ACE2 and SLC6A19 in the cecum and levels of Gly, Ser, Gln, and Trp in the serum similar to that of uninfected chicks (P < 0.05). Additionally, E. coli Nissle 1917 altered cecum microbiota composition and enriched the abundance of E. coli, Lactobacillales, and Lachnospiraceae. These findings reveal that the probiotic E. coli Nissle 1917 reduced S. Enteritidis infection and shows enormous potential as an alternative to antibiotics.
Collapse
Affiliation(s)
| | | | - Guanglei Cong
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Yunqi Xiao
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Yiru Shen
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Shan Zhang
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Wenchang Zhao
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Shourong Shi
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| |
Collapse
|
16
|
Wu Y, Hu A, Shu X, Huang W, Zhang R, Xu Y, Yang C. Lactobacillus plantarum postbiotics trigger AMPK-dependent autophagy to suppress Salmonella intracellular infection and NLRP3 inflammasome activation. J Cell Physiol 2023; 238:1336-1353. [PMID: 37052047 DOI: 10.1002/jcp.31016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
We previously found that Lactobacillus plantarum (LP)-derived postbiotics protected animals against Salmonella infection, but the molecular mechanism remains obscure. This study clarified the mechanisms from the perspective of autophagy. Intestinal porcine epithelial cells (IPEC-J2) were pretreated with LP-derived postbiotics (the culture supernatant, LPC; or heat-killed bacteria, LPB), and then challenged with Salmonella enterica Typhimurium (ST). Results showed that LP postbiotics markedly triggered autophagy under ST infection, as indicated by the increased LC3 and Beclin1 and the decreased p62 levels. Meanwhile, LP postbiotics (particularly LPC) exhibited a strong capacity of inhibiting ST adhesion, invasion and replication. Pretreatment with the autophagy inhibitor 3-methyladenine (3-MA) led to a significant decrease of autophagy and the aggravated infection, indicating the importance of autophagy in LP postbiotics-mediated Salmonella elimination. LP postbiotics (especially LPB) significantly suppressed ST-induced inflammation by modulating inflammatory cytokines (the increased interleukin (IL)-4 and IL-10, and decreased tumor necrosis factor-α (TNF), IL-1β, IL-6 and IL-18). Furthermore, LP postbiotics inhibited NOD-like receptor protein 3 (NLRP3) inflammasome activation, as evidenced by the decreased levels of NLRP3, Caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). Deficits in autophagy resulted in an increase of inflammatory response and inflammasome activation. Finally, we found that both LPC and LPB triggered AMP-activated protein kinase (AMPK) signaling pathway to induce autophagy, and this was further confirmed by AMPK RNA interference. The intracellular infection and NLRP3 inflammasome were aggravated after AMPK knockdown. In summary, LP postbiotics trigger AMPK-mediated autophagy to suppress Salmonella intracellular infection and NLRP3 inflammasome in IPEC-J2 cells. Our findings highlight the effectiveness of postbiotics, and provide a new strategy for preventing Salmonella infection.
Collapse
Affiliation(s)
- Yanping Wu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Aixin Hu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Xin Shu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Wenxia Huang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Ruiqiang Zhang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Yinglei Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Caimei Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| |
Collapse
|
17
|
Tian L, Zhong C, He Y, Lu Q, Wang Y, Zhao X, Wei H, Tao X. Preventive of Lacticaseibacillus casei WLCA02 against Salmonella Typhimurium infection via strengthening the intestinal barrier and activating the macrophages. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
18
|
Albuquerque Pereira MDF, Morais de Ávila LG, Dos Santos Cruz BC, Campos Silva B, Licursi de Oliveira L, Vilela Gonçalves R, de Oliveira Barros Ribon A, de Oliveira Mendes TA, Gouveia Peluzio MDC. The role of IL-10 in regulating inflammation and gut microbiome in mice consuming milk kefir and orally challenged with S. Typhimurium. Food Funct 2023; 14:3804-3814. [PMID: 37000521 DOI: 10.1039/d2fo04063h] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
Kefir has been suggested as a possible bacterial prophylaxis against Salmonella and IL-10 production seems to be crucial in the pathogenesis of salmonellosis in mice. This study evaluated the role of IL-10 in the inflammation and gut microbiome in mice consuming milk kefir and orally challenged with Salmonella enterica serovar Typhimurium. C57BL wild type (WT) (n = 40) and C57BL IL-10-/- (KO) (n = 40) mice were subdivided into eight experimental groups either treated or not with kefir. In the first 15 days, the water groups received filtered water (0.1 mL) while the kefir groups received milk kefir (10% w/v) orally by gavage. Then, two groups of each strain received a single dose (0.1 mL) of the inoculum of S. Typhimurium (ATCC 14028, dose: 106 CFU mL-1). After four weeks, the animals were euthanized to remove the colon for further analysis. Kefir prevented systemic infections only in IL-10-/- mice, which were able to survive, regulate cytokines, and control colon inflammation. The abundance in Lachnospiraceae and Roseburia, and also the higher SCFA production in the pre-infection, showed that kefir has a role in intestinal health and protection, colonizing and offering competition for nutrients with the pathogen as well as acting in the regulation of salmonella infectivity only in the absence of IL-10. These results demonstrate the role of IL-10 in the prognosis of salmonellosis and how milk kefir can be used in acute infections.
Collapse
Affiliation(s)
| | - Larissa Gabriela Morais de Ávila
- Interunit Postgraduate Program in Bioinformatics, Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, Minas Gerais, Brazil.
| | - Bruna Cristina Dos Santos Cruz
- Biological Sciences and Health Institute, Rodovia BR230 KM 7, Rio Paranaíba - MG, Universidade Federal de Viçosa, Brazil.
| | - Bruno Campos Silva
- Department of Biochemistry and Molecular Biology, Universidade Federal de Viçosa - UFV, Viçosa, Minas Gerais, Brazil.
| | | | | | | | | | | |
Collapse
|
19
|
Liu W, Huang Y, Zhang H, Liu Z, Huan Q, Xiao X, Wang Z. Factors and Mechanisms Influencing Conjugation In Vivo in the Gastrointestinal Tract Environment: A Review. Int J Mol Sci 2023; 24:5919. [PMID: 36982992 PMCID: PMC10059276 DOI: 10.3390/ijms24065919] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The emergence and spread of antibiotic resistance genes (ARGs) have imposed a serious threat on global public health. Horizontal gene transfer (HGT) via plasmids is mainly responsible for the spread of ARGs, and conjugation plays an important role in HGT. The conjugation process is very active in vivo and its effect on the spreading of ARGs may be underestimated. In this review, factors affecting conjugation in vivo, especially in the intestinal environment, are summarized. In addition, the potential mechanisms affecting conjugation in vivo are summarized from the perspectives of bacterial colonization and the conjugation process.
Collapse
Affiliation(s)
- Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Yanhu Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Han Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Ziyi Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Quanmin Huan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Xia Xiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225012, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225012, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou 225012, China
| |
Collapse
|
20
|
Mistry JJ, Bowles K, Rushworth SA. HSC-derived fatty acid oxidation in steady-state and stressed hematopoiesis. Exp Hematol 2023; 117:1-8. [PMID: 36223830 DOI: 10.1016/j.exphem.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 10/04/2022] [Indexed: 01/10/2023]
Abstract
Metabolism impacts all cellular functions and plays a fundamental role in physiology. Metabolic regulation of hematopoiesis is dynamically regulated under steady-state and stress conditions. It is clear that hematopoietic stem cells (HSCs) impose different energy demands and flexibility during maintenance compared with stressed conditions. However, the cellular and molecular mechanisms underlying metabolic regulation in HSCs remain poorly understood. In this review, we focus on defining the role of fatty acid oxidation (FAO) in HSCs. We first review the existing literature describing FAO in HSCs under steady-state hematopoiesis. Next, we describe the models used to examine HSCs under stress conditions, and, finally, we describe how infection causes a shift toward FAO in HSCs and the impact of using this pathway on emergency hematopoiesis.
Collapse
Affiliation(s)
| | - Kristian Bowles
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom; Department of Haematology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Stuart A Rushworth
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom.
| |
Collapse
|
21
|
Wang Z, Zhu S, Li C, Lyu L, Yu J, Wang D, Xu Z, Ni J, Gao B, Lu J, Yao YF. Gene essentiality profiling reveals a novel determinant of stresses preventing protein aggregation in Salmonella. Emerg Microbes Infect 2022; 11:1554-1571. [PMID: 35603550 PMCID: PMC9176671 DOI: 10.1080/22221751.2022.2081618] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Adaptation to various stresses during infection is important for Salmonella Typhimurium virulence, while the fitness determinants under infection-relevant stress conditions remain unknown. Here, we simulated conditions Salmonella encountered within the host or in the environment by 15 individual stresses as well as two model cell lines (epithelium and macrophage) to decipher the genes and pathways required for fitness. By high-resolution Tn-seq analysis, a total of 1242 genes were identified as essential for fitness under at least one stress condition. The comparative analysis of fitness determinants in 17 stress conditions indicated the essentiality of genes varied in different mimicking host niches. A total of 12 genes were identified as fitness determinants in all stress conditions, including recB, recC, and xseA (encode three exonuclease subunits necessary for DNA recombination repair) and a novel essential fitness gene yheM. YheM is a putative sulfurtransferase subunit that is responsible for tRNA modification, and our results showed that Salmonella lacking yheM accumulated more aggregates of endogenous protein than wild-type. Moreover, we established a scoring scheme for sRNA essentiality analysis and found STnc2080 of unknown function was essential for resistance to LL-37. In summary, we systematically dissected Salmonella gene essentiality profiling and demonstrated the general and specific adaptive requirements in infection-relevant niches. Our data not only provide valuable insights on how Salmonella responds to environmental stresses during infections but also highlight the potential clinical application of fitness determinants in vaccine development.
Collapse
Affiliation(s)
- Zuoqiang Wang
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Siqi Zhu
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Congcong Li
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lin Lyu
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jingchen Yu
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhihong Xu
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Beile Gao
- CAS Key Laboratory of Tropical Marine Bio Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, People's Republic of China.,Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, People's Republic of China
| | - Jie Lu
- Department of Infectious Diseases, Shanghai Ruijin Hospital, Shanghai, People's Republic of China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Department of Infectious Diseases, Shanghai Ruijin Hospital, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Ghasemi A, Wang S, Sahay B, Abbott JR, Curtiss R. Protective immunity enhanced Salmonella vaccine vectors delivering Helicobacter pylori antigens reduce H. pylori stomach colonization in mice. Front Immunol 2022; 13:1034683. [PMID: 36466847 PMCID: PMC9716130 DOI: 10.3389/fimmu.2022.1034683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Helicobacter pylori is a major cause of gastric mucosal inflammation, peptic ulcers, and gastric cancer. Emerging antimicrobial-resistant H. pylori has hampered the effective eradication of frequent chronic infections. Moreover, a safe vaccine is highly demanded due to the absence of effective vaccines against H. pylori. In this study, we employed a new innovative Protective Immunity Enhanced Salmonella Vaccine (PIESV) vector strain to deliver and express multiple H. pylori antigen genes. Immunization of mice with our vaccine delivering the HpaA, Hp-NAP, UreA and UreB antigens, provided sterile protection against H. pylori SS1 infection in 7 out of 10 tested mice. In comparison to the control groups that had received PBS or a PIESV carrying an empty vector, immunized mice exhibited specific and significant cellular recall responses and antigen-specific serum IgG1, IgG2c, total IgG and gastric IgA antibody titers. In conclusion, an improved S. Typhimurium-based live vaccine delivering four antigens shows promise as a safe and effective vaccine against H. pylori infection.
Collapse
Affiliation(s)
- Amir Ghasemi
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Jeffrey R. Abbott
- Department of Comparative, Diagnostic and Population Medicine, University of Florida, Gainesville, FL, United States
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| |
Collapse
|
23
|
Sedivy-Haley K, Blimkie T, Falsafi R, Lee AHY, Hancock REW. A transcriptomic analysis of the effects of macrophage polarization and endotoxin tolerance on the response to Salmonella. PLoS One 2022; 17:e0276010. [PMID: 36240188 PMCID: PMC9565388 DOI: 10.1371/journal.pone.0276010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella is an intracellular pathogen causing significant morbidity and mortality. Its ability to grow inside macrophages is important to virulence, and is dependent on the activation state of the macrophages. Classically activated M1 macrophages are non-permissive for Salmonella growth, while alternatively activated M2 macrophages are permissive for Salmonella growth. Here we showed that endotoxin-primed macrophages (MEP), such as those associated with sepsis, showed similar levels of Salmonella resistance to M1 macrophages after 2 hr of intracellular infection, but at the 4 hr and 24 hr time points were susceptible like M2 macrophages. To understand this mechanistically, transcriptomic sequencing, RNA-Seq, was performed. This showed that M1 and MEP macrophages that had not been exposed to Salmonella, demonstrated a process termed here as primed activation, in expressing relatively higher levels of particular anti-infective genes and pathways, including the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway. In contrast, in M2 macrophages these genes and pathways were largely expressed only in response to infection. Conversely, in response to infection, M1 macrophages, but not MEP macrophages, modulated additional genes known to be associated with susceptibility to Salmonella infection, possibly contributing to the differences in resistance at later time points. Application of the JAK inhibitor Ruxolitinib before infection reduced resistance in M1 macrophages, supporting the importance of early JAK-STAT signalling in M1 resistance to Salmonella.
Collapse
Affiliation(s)
- Katharine Sedivy-Haley
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
24
|
Agbayani G, Clark K, Sad S, Murphy SP, Krishnan L. Roles of natural resistance-associated macrophage protein-1 in modulating bacterial distribution and immune responses during Salmonella enterica serovar Typhimurium infection in murine pregnancy. Am J Reprod Immunol 2022; 88:e13599. [PMID: 35851978 PMCID: PMC9509426 DOI: 10.1111/aji.13599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/23/2022] [Accepted: 07/05/2022] [Indexed: 11/27/2022] Open
Abstract
PROBLEM Salmonella enterica serovar Typhimurium (S.Tm) infection in Nramp1+/+ mice during pregnancy can lead to profound bacterial growth in the feto-placental unit and adverse pregnancy outcomes, including fetal loss, maternal illness and death. The kinetics and mechanisms by which S.Tm gains entry within individual feto-placental unit, and disseminates through tissues leading to placental resorption and fetal demise remain unclear. METHOD OF STUDY Mice were systemically infected with S.Tm. Bacterial burden within spleen and individual placentas, and placental/fetal resorptions were quantified. Flow cytometric analysis of immune cell types in the spleen and individual placentas was performed. Cytokine expression in maternal serum was determined through cytometric bead array. RESULTS Systemic infection with S.Tm resulted in preferential bacterial proliferation in placentas compared to the spleen in Nramp1+/+ mice. At 24 h post-infection, the mean infection rate of individual placentas per mouse was ∼50%, increasing to >75% by 72 h post-infection, suggesting that initial infection in few sites progresses to rapid spread of infection through the uterine milieu. This correlated with a steady increase in placental/fetal resorption rates. Placental infection was associated with local increased neutrophil percentages, whereas numbers and percentages in the spleen remained unchanged, suggesting dichotomous modulation of inflammation between the systemic compartment and the feto-maternal interface. Reduced survival rates of pregnant mice during infection correlated with decreased serum IFN-γ but increased IL-10 levels relative to non-pregnant controls. CONCLUSION Pregnancy compromises host resistance conferred by Nramp1 against S.Tm through compartment-specific regulation of maternal and placental cellular responses, and modulation of systemic cytokine expression.
Collapse
Affiliation(s)
- Gerard Agbayani
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, Ontario, Canada
| | - Kristina Clark
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, Ontario, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Shawn P Murphy
- Department of Obstetrics and Gynecology, University of Rochester, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Lakshmi Krishnan
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, Ontario, Canada
| |
Collapse
|
25
|
Ding C, Wu H, Cao X, Gao Z, Tang Z, Fan W, Yan L, Liu B, Lin H, Song S. Lactobacillus crispatus-derived exopolysaccharides with antibacterial activity limit Salmonella typhimurium invasion by inhibiting inflammasome-mediated pyroptosis. Food Funct 2022; 13:10501-10515. [PMID: 36148688 DOI: 10.1039/d2fo02125k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a novel heteropolysaccharide (EPS 7-4) with a molecular weight of 53 387 Da was isolated from Lactobacillus crispatus, and it was mainly composed of mannose (36.9%) and glucose (30.8%). EPS 7-4 showed excellent inhibitory effects on the proliferation, biofilm formation, and virulence factor gene expression of Salmonella typhimurium (S. typhimurium) by disrupting the integrity of the bacterial wall. Furthermore, EPS 7-4 can effectively restrict bacterial translocation, upregulate the abundance of Lactobacillus spp. and Bifidobacterium spp., and alleviate the S. typhimurium induced severe inflammatory response in the intestinal tract of mice. Besides, we demonstrated that EPS 7-4 can protect mice by inhibiting S. typhimurium induced pyroptosis, with the mechanism that EPS 7-4 affects ASC oligomerization during inflammasome-mediated pyroptosis. Therefore, due to its excellent anti-bacterial and anti-inflammatory abilities, EPS 7-4 is a promising health regulator owing to its excellent antibacterial and anti-inflammatory abilities.
Collapse
Affiliation(s)
- Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Huixian Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China. .,Management Office of Dafeng Milu National Nature Reserve, Yancheng, 224136, China
| | - Xiuyun Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Zhihui Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Liping Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Bin Liu
- Management Office of Dafeng Milu National Nature Reserve, Yancheng, 224136, China
| | - Hong Lin
- Animal, Plant and Food Inspection Center, Nanjing Customs, Nanjing, 210019, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
26
|
Zhang G, Wang J, Zhao Z, Xin T, Fan X, Shen Q, Raheem A, Lee CR, Jiang H, Ding J. Regulated necrosis, a proinflammatory cell death, potentially counteracts pathogenic infections. Cell Death Dis 2022; 13:637. [PMID: 35869043 PMCID: PMC9307826 DOI: 10.1038/s41419-022-05066-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023]
Abstract
Since the discovery of cell apoptosis, other gene-regulated cell deaths are gradually appreciated, including pyroptosis, ferroptosis, and necroptosis. Necroptosis is, so far, one of the best-characterized regulated necrosis. In response to diverse stimuli (death receptor or toll-like receptor stimulation, pathogenic infection, or other factors), necroptosis is initiated and precisely regulated by the receptor-interacting protein kinase 3 (RIPK3) with the involvement of its partners (RIPK1, TRIF, DAI, or others), ultimately leading to the activation of its downstream substrate, mixed lineage kinase domain-like (MLKL). Necroptosis plays a significant role in the host's defense against pathogenic infections. Although much has been recognized regarding modulatory mechanisms of necroptosis during pathogenic infection, the exact role of necroptosis at different stages of infectious diseases is still being unveiled, e.g., how and when pathogens utilize or evade necroptosis to facilitate their invasion and how hosts manipulate necroptosis to counteract these detrimental effects brought by pathogenic infections and further eliminate the encroaching pathogens. In this review, we summarize and discuss the recent progress in the role of necroptosis during a series of viral, bacterial, and parasitic infections with zoonotic potentials, aiming to provide references and directions for the prevention and control of infectious diseases of both human and animals.
Collapse
Affiliation(s)
- Guangzhi Zhang
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Jinyong Wang
- grid.508381.70000 0004 0647 272XShenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen, 518000 China ,grid.258164.c0000 0004 1790 3548Institute of Respiratory Diseases, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, 518020 Guangdong China
| | - Zhanran Zhao
- grid.47840.3f0000 0001 2181 7878Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, CA 94720-3200 USA
| | - Ting Xin
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Xuezheng Fan
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Qingchun Shen
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Abdul Raheem
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China ,grid.35155.370000 0004 1790 4137Present Address: Huazhong Agricultural University, Wuhan, China
| | - Chae Rhim Lee
- grid.47840.3f0000 0001 2181 7878Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, CA 94720-3200 USA ,grid.266093.80000 0001 0668 7243Present Address: University of California, Irvine, CA USA
| | - Hui Jiang
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Jiabo Ding
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| |
Collapse
|
27
|
Liu Z, Zhu S, He M, Li M, Wei H, Zhang L, Sun Q, Jia Q, Hu N, Fang Y, Song L, Zhou C, Tao H, Kao JY, Zhu H, Owyang C, Duan L. Patients with breath test positive are necessary to be identified from irritable bowel syndrome: a clinical trial based on microbiomics and rifaximin sensitivity. Chin Med J (Engl) 2022; 135:1716-1727. [PMID: 36070467 PMCID: PMC9509105 DOI: 10.1097/cm9.0000000000002294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND As a non-invasive and effective diagnostic method for small intestinal bacterial overgrowth (SIBO), wild-use of breath test (BT) has demonstrated a high comorbidity rate in patients with diarrhea-predominant irritable bowel syndrome (IBS-D) and SIBO. Patients overlapping with SIBO respond better to rifaximin therapy than those with IBS-D only. Gut microbiota plays a critical role in both of these two diseases. We aimed to determine the microbial difference between IBS-D overlapping with/without SIBO, and to study the underlying mechanism of its sensitivity to rifaximin. METHODS Patients with IBS-D were categorized as BT-negative (IBSN) and BT-positive (IBSP). Healthy volunteers (BT-negative) were enrolled as healthy control. The patients were clinically evaluated before and after rifaximin treatment (0.4 g bid, 4 weeks). Blood, intestine, and stool samples were collected for cytokine assessment and gut microbial analyses. RESULTS Clinical complaints and microbial abundance were significantly higher in IBSP than in IBSN. In contrast, severe systemic inflammation and more active bacterial invasion function that were associated with enrichment of opportunistic pathogens were seen in IBSN. The symptoms of IBSP patients were relieved in different degrees after therapy, but the symptoms of IBSN rarely changed. We also found that the presence of IBSN-enriched genera ( Enterobacter and Enterococcus ) are unaffected by rifaximin therapy. CONCLUSIONS IBS-D patients overlapping with SIBO showed noticeably different fecal microbial composition and function compared with IBS-D only. The better response to rifaximin in those comorbid patients might associate with their different gut microbiota, which suggests that BT is necessary before IBS-D diagnosis and use of rifaximin. REGISTRATION Chinese Clinical Trial Registry, ChiCTR1800017911.
Collapse
Affiliation(s)
- Zuojing Liu
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Shiwei Zhu
- Department of Ultrasound, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Meibo He
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mo Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100187, China
| | - Hui Wei
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Lu Zhang
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Qinghua Sun
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Qiong Jia
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Nan Hu
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Yuan Fang
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Lijin Song
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Chen Zhou
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - Heqing Tao
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| | - John Y Kao
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, 6520 MSRB I, SPC 5682, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Huaiqiu Zhu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100187, China
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, 6520 MSRB I, SPC 5682, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Haidian District, Beijing 100191, China
| |
Collapse
|
28
|
Oral Administration with Recombinant Attenuated Regulated Delayed Lysis Salmonella Vaccines Protecting against Staphylococcus aureus Kidney Abscess Formation. Vaccines (Basel) 2022; 10:vaccines10071073. [PMID: 35891237 PMCID: PMC9324569 DOI: 10.3390/vaccines10071073] [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: 05/10/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 12/10/2022] Open
Abstract
Abscess formation is one of the main symptoms of Staphylococcus aureus infection. It is very important to inhibit abscess formation for preventing S. aureus persistent infection. To find a feasible solution, the live oral vaccines delivering S. aureus antigens, rEsxAB and rHlam, were constructed, which were based on the attenuated regulated delayed lysis Salmonella enterica subspecies Serovar Typhimurium strain χ11802, and the inhibiting effect on abscess formation was evaluated in mice kidneys. As the results showed, after oral administration, humoral immunity was induced via the mucosal route as the antigen-specific IgG in the serum and IgA in the intestinal mucus both showed significant increases. Meanwhile, the production of IFN-γ and IL-17 in the kidney tissue suggested that Th1/Th17-biased cellular immunity played a role in varying degrees. After challenged intravenously (i.v.) with S. aureus USA300, the χ11802(pYA3681−esxAB)-vaccinated group showed obvious inhibition in kidney abscess formation among the vaccinated group, as the kidney abscess incidence rate and the staphylococcal load significantly reduced, and the kidney pathological injury was improved significantly. In conclusion, this study provided experimental data and showed great potential for live oral vaccine development with the attenuated regulated delayed lysis Salmonella Typhimurium strains against S. aureus infection.
Collapse
|
29
|
Herrera-Uribe J, Zaldívar-López S, Aguilar C, Entrenas-García C, Bautista R, Claros MG, Garrido JJ. Study of microRNA expression in Salmonella Typhimurium-infected porcine ileum reveals miR-194a-5p as an important regulator of the TLR4-mediated inflammatory response. Vet Res 2022; 53:35. [PMID: 35598011 PMCID: PMC9123658 DOI: 10.1186/s13567-022-01056-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Infection with Salmonella Typhimurium (S. Typhimurium) is a common cause of food-borne zoonosis leading to acute gastroenteritis in humans and pigs, causing economic losses to producers and farmers, and generating a food security risk. In a previous study, we demonstrated that S. Typhimurium infection produces a severe transcriptional activation of inflammatory processes in ileum. However, little is known regarding how microRNAs regulate this response during infection. Here, small RNA sequencing was used to identify 28 miRNAs differentially expressed (DE) in ileum of S. Typhimurium-infected pigs, which potentially regulate 14 target genes involved in immune system processes such as regulation of cytokine production, monocyte chemotaxis, or cellular response to interferon gamma. Using in vitro functional and gain/loss of function (mimics/CRISPR-Cas system) approaches, we show that porcine miR-194a-5p (homologous to human miR-194-5p) regulates TLR4 gene expression, an important molecule involved in pathogen virulence, recognition and activation of innate immunity in Salmonella infection.
Collapse
Affiliation(s)
- Juber Herrera-Uribe
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Viral Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain. .,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain.
| | - Carmen Aguilar
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Carmen Entrenas-García
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain
| | - Rocío Bautista
- Andalusian Platform of Bioinformatics-SCBI, University of Málaga, Málaga, Spain
| | - M Gonzalo Claros
- Andalusian Platform of Bioinformatics-SCBI, University of Málaga, Málaga, Spain.,Department of Molecular Biology and Biochemistry, University of Málaga, Málaga, Spain
| | - Juan J Garrido
- Immunogenomics and Molecular Pathogenesis Group, Department of Genetics, Faculty of Veterinary Medicine, University of Córdoba, Córdoba, Spain.,Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Research Group GA-14, Córdoba, Spain
| |
Collapse
|
30
|
Ma Y, Khan MZ, Xiao J, Alugongo GM, Chen X, Li S, Wang Y, Cao Z. An Overview of Waste Milk Feeding Effect on Growth Performance, Metabolism, Antioxidant Status and Immunity of Dairy Calves. Front Vet Sci 2022; 9:898295. [PMID: 35656173 PMCID: PMC9152456 DOI: 10.3389/fvets.2022.898295] [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: 03/20/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Waste milk (WM) is a part of the milk produced on dairy farms, which is usually unsuitable for human consumption. The WM contains transition milk, mastitis milk, colostrum, milk with somatic cells, blood (Hemolactia), harmful pathogens, pathogenic and antibiotic residues. Due to the high cost of milk replacer (MR), dairy farmers prefer raw WM to feed their calves. It has been well established that WM has a greater nutritive value than MR. Hence WM can contribute to improved growth, rumen development, and immune-associated parameters when fed to dairy calves. However, feeding raw WM before weaning has continuously raised some critical concerns. The pathogenic load and antibiotic residues in raw WM may increase the risk of diseases and antibacterial resistance in calves. Thus, pasteurization has been recommended as an effective method to decrease the risk of diseases in calves by killing/inhibiting the pathogenic microorganisms in the raw WM. Altogether, the current review provides a brief overview of the interplay between the positive role of raw WM in the overall performance of dairy calves, limitations of raw WM as a feed source and how to overcome these issues arising from feeding raw WM.
Collapse
Affiliation(s)
- Yulin Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Muhammad Zahoor Khan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- University of Agriculture, Dera Ismail Khan, Khyber Pakhtunkhwa, Pakistan
| | - Jianxin Xiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Gibson Maswayi Alugongo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xu Chen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Zhijun Cao
| |
Collapse
|
31
|
Antimicrobial and Immunomodulatory Effects of Selected Chemokine and Antimicrobial Peptide on Cytokine Profile during Salmonella Typhimurium Infection in Mouse. Antibiotics (Basel) 2022; 11:antibiotics11050607. [PMID: 35625251 PMCID: PMC9137564 DOI: 10.3390/antibiotics11050607] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
The antimicrobial and immunomodulatory capacities of the peptide Css54 and the chemokine MCP-1 were tested. The first, a peptide isolated from the venom of the scorpion Centruroides suffusus suffusus was synthesized chemically. In contrast, the second is a monocyte chemoattractant expressed as a recombinant protein in our lab. It was observed in vitro that Css54 inhibited the growth of Salmonella enterica serovar Typhimurium (6.2 µg/mL). At high concentrations, it was toxic to macrophages (25 µg/mL), activated macrophage phagocytosis (1.5 µg/mL), and bound Salmonella LPS (3 µg/mL). On the other hand, the recombinant MCP-1 neither inhibited the growth of Salmonella Typhimurium nor was it toxic to macrophages (up to 25 µg/mL), nor activated macrophage phagocytosis or bound Salmonella LPS (up to 3 µg/mL). Although it was observed in vivo in mice Balb/C that both Css54 and MCP-1 did not resolve the intraperitoneal infection by S. Typhimurium, Css54 decreased the expression of IL-6 and increased IL-10, IL-12p70, and TNF-α levels; meanwhile, MCP-1 decreased the expression of IFN-γ and increased IL-12p70 and TNF-α. It was also observed that the combination of both molecules Css54 and MCP-1 increased the expression of IL-10 and TNF-α.
Collapse
|
32
|
Devlin JR, Santus W, Mendez J, Peng W, Yu A, Wang J, Alejandro-Navarreto X, Kiernan K, Singh M, Jiang P, Mechref Y, Behnsen J. Salmonella enterica serovar Typhimurium chitinases modulate the intestinal glycome and promote small intestinal invasion. PLoS Pathog 2022; 18:e1010167. [PMID: 35482787 PMCID: PMC9049507 DOI: 10.1371/journal.ppat.1010167] [Citation(s) in RCA: 5] [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: 12/03/2021] [Accepted: 03/23/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is one of the leading causes of food-borne illnesses worldwide. To colonize the gastrointestinal tract, S. Typhimurium produces multiple virulence factors that facilitate cellular invasion. Chitinases have been recently emerging as virulence factors for various pathogenic bacterial species, and the S. Typhimurium genome contains two annotated chitinases: STM0018 (chiA) and STM0233. However, the role of these chitinases during S. Typhimurium pathogenesis is unknown. The putative chitinase STM0233 has not been studied previously, and only limited data exists on ChiA. Chitinases typically hydrolyze chitin polymers, which are absent in vertebrates. However, chiA expression was detected in infection models and purified ChiA cleaved carbohydrate subunits present on mammalian surface glycoproteins, indicating a role during pathogenesis. Here, we demonstrate that expression of chiA and STM0233 is upregulated in the mouse gut and that both chitinases facilitate epithelial cell adhesion and invasion. S. Typhimurium lacking both chitinases showed a 70% reduction in invasion of small intestinal epithelial cells in vitro. In a gastroenteritis mouse model, chitinase-deficient S. Typhimurium strains were also significantly attenuated in the invasion of small intestinal tissue. This reduced invasion resulted in significantly delayed S. Typhimurium dissemination to the spleen and the liver, but chitinases were not required for systemic survival. The invasion defect of the chitinase-deficient strain was rescued by the presence of wild-type S. Typhimurium, suggesting that chitinases are secreted. By analyzing N-linked glycans of small intestinal cells, we identified specific N-acetylglucosamine-containing glycans as potential extracellular targets of S. Typhimurium chitinases. This analysis also revealed a differential abundance of Lewis X/A-containing glycans that is likely a result of host cell modulation due to the detection of S. Typhimurium chitinases. Similar glycomic changes elicited by chitinase deficient strains indicate functional redundancy of the chitinases. Overall, our results demonstrate that S. Typhimurium chitinases contribute to intestinal adhesion and invasion through modulation of the host glycome.
Collapse
Affiliation(s)
- Jason R. Devlin
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - William Santus
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Jorge Mendez
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Wenjing Peng
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Aiying Yu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Junyao Wang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Xiomarie Alejandro-Navarreto
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Kaitlyn Kiernan
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Manmeet Singh
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Peilin Jiang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Judith Behnsen
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
33
|
Zhao C, Chen H, Liang H, Zhao X, Tang W, Wei M, Li Y, Zhang J, Yu X, Chen G, Zhu H, Jiang L, Zhang X. Lactobacillus plantarum RS-09 Induces M1-Type Macrophage Immunity Against Salmonella Typhimurium Challenge via the TLR2/NF-κB Signalling Pathway. Front Pharmacol 2022; 13:832245. [PMID: 35355723 PMCID: PMC8959098 DOI: 10.3389/fphar.2022.832245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/26/2022] [Indexed: 12/30/2022] Open
Abstract
Lactobacillus plantarum can interact with macrophages against bacterial enteropathy due to its potential ability to modulate macrophage polarization. However, this mechanism is not completely understood. TLR2 can recognize microbial components and trigger macrophage cytokine responses to different gram-positive strains. The aim of this study was to investigate whether probiotic Lactobacillus plantarum RS-09 can induce macrophage polarization against Salmonella Typhimurium infection via TLR2 signalling. BALB/c mice were preadministered RS-09 continuously for 7 days and then infected with Salmonella Typhimurium ATCC14028. Mouse RAW264.7 mononuclear macrophages were stimulated with RS-09 and coincubated with ATCC14028 or PBS controls. The results of the in vivo study indicated that RS-09 could relieve S. Typhimurium-induced splenomegaly, body weight loss and death rate. RS-09 also limited the colonization and translocation of S. Typhimurium in the gastrointestinal tract and thereby protected against infection. We also observed that RS-09 upregulated the production of M1 macrophage characteristics (e.g., CD11c and IL-6) against S. Typhimurium. Furthermore, RS-09 induced the expression of TLR2 in macrophages. In an in vitro study, treatment of RAW264.7 cells with RS-09 either concurrently with or before S. Typhimurium challenge enhanced the secretion of Reactive oxygen species and Nitric oxide. This effect was related to TLR2 and NF-κB activation. Based on these findings, Lactobacillus plantarum RS-09 was shown to modulate M1 macrophage polarization and induce TLR2-linked NF-κB signalling activity in the innate immune response to S. Typhimurium infection.
Collapse
Affiliation(s)
- Chenpei Zhao
- School of Life Sciences, Ludong University, Yantai, China
| | - Huan Chen
- School of Life Sciences, Ludong University, Yantai, China
| | - Hao Liang
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyu Zhao
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Wenli Tang
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Maolian Wei
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Youzhi Li
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Xin Yu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
| | - Guozhong Chen
- School of Life Sciences, Ludong University, Yantai, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Linlin Jiang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| |
Collapse
|
34
|
Garcia Gonzalez J, Hernandez FJ. Nuclease activity: an exploitable biomarker in bacterial infections. Expert Rev Mol Diagn 2022; 22:265-294. [PMID: 35240900 DOI: 10.1080/14737159.2022.2049249] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION In the increasingly challenging field of clinical microbiology, diagnosis is a cornerstone whose accuracy and timing are crucial for the successful management, therapy, and outcome of infectious diseases. Currently employed biomarkers of infectious diseases define the scope and limitations of diagnostic techniques. As such, expanding the biomarker catalog is crucial to address unmet needs and bring about novel diagnostic functionalities and applications. AREAS COVERED This review describes the extracellular nucleases of 15 relevant bacterial pathogens and discusses the potential use of nuclease activity as a diagnostic biomarker. Articles were searched for in PubMed using terms: "nuclease", "bacteria", "nuclease activity" or "biomarker". For overview sections, original and review articles between 2000 and 2019 were searched for using terms: "infections", "diagnosis", "bacterial", "burden", "challenges". Informative articles were selected. EXPERT OPINION Using the catalytic activity of nucleases offers new possibilities compared to established biomarkers. Nucleic acid activatable reporters in combination with different transduction platforms and delivery methods can be used to detect disease-associated nuclease activity patterns in vitro and in vivo for prognostic and diagnostic applications. Even when these patterns are not obvious or of unknown etiology, screening platforms could be used to identify new disease reporters.
Collapse
Affiliation(s)
- Javier Garcia Gonzalez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| | - Frank J Hernandez
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Wallenberg Centre for Molecular Medicine (WCMM), Linköping, Sweden.,Nucleic Acids Technologies Laboratory (NAT-lab), Linköping University, Linköping, Sweden
| |
Collapse
|
35
|
Cui C, Liu Q, Duan B, Liu X, Wei H, Peng J. Bioactive triple peptide inhibits inflammasome activation to alleviate Salmonella-induced intestinal inflammation in mice via modulation of host defense and bacterial virulence. Food Funct 2022; 13:3512-3525. [DOI: 10.1039/d1fo03891e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Over the past long period, Salmonella Typhimurium has been an important pathogen that causes intestinal diseases and spells enormous economic shock to animal husbandry all over the world. Pyroptosis and...
Collapse
|
36
|
Kotas ME, Mroz NM, Koga S, Liang HE, Schroeder AW, Ricardo-Gonzalez RR, Schneider C, Locksley RM. CISH constrains the tuft-ILC2 circuit to set epithelial and immune tone. Mucosal Immunol 2021; 14:1295-1305. [PMID: 34290377 PMCID: PMC8528700 DOI: 10.1038/s41385-021-00430-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/15/2021] [Accepted: 07/04/2021] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILCs) are tissue-resident effectors poised to activate rapidly in response to local signals such as cytokines. To preserve homeostasis, ILCs must employ multiple pathways, including tonic suppressive mechanisms, to regulate their primed state and prevent inappropriate activation and immunopathology. Such mechanisms remain incompletely characterized. Here we show that cytokine-inducible SH2-containing protein (CISH), a suppressor of cytokine signaling (SOCS) family member, is highly and constitutively expressed in type 2 innate lymphoid cells (ILC2s). Mice that lack CISH either globally or conditionally in ILC2s show increased ILC2 expansion and activation, in association with reduced expression of genes inhibiting cell-cycle progression. Augmented proliferation and activation of CISH-deficient ILC2s increases basal and inflammation-induced numbers of intestinal tuft cells and accelerates clearance of the model helminth, Nippostrongylus brasiliensis, but compromises innate control of Salmonella typhimurium. Thus, CISH constrains ILC2 activity both tonically and after perturbation, and contributes to the regulation of immunity in mucosal tissue.
Collapse
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy & Sleep Medicine, University of California, San Francisco, CA, USA
| | - Nicholas M Mroz
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Satoshi Koga
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Hong-Erh Liang
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | | | | | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.
| |
Collapse
|
37
|
Li WH, Liu YL, Lun JC, He YM, Tang LP. Heat stress inhibits TLR4-NF-κB and TLR4-TBK1 signaling pathways in broilers infected with Salmonella Typhimurium. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2021; 65:1895-1903. [PMID: 34061266 PMCID: PMC8536552 DOI: 10.1007/s00484-021-02146-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 05/29/2023]
Abstract
With the global warming, the harm of heat stress (HS) to the breeding industry has become more common, which causes the decline of animal production performance and low immunity. This study aimed to analyze the effect of HS on the intestinal immune function of Salmonella-infected chickens. Fourteen-day-old broilers were divided into the following four groups of eight replicates: control (Control), heat stress (HS), Salmonella Typhimurium (ST), and heat stress + Salmonella Typhimurium (HS+ST). The broilers were subjected to a heat stress of 35 °C from 15 to 28 days of age. Salmonella Typhimurium (ST, 14028, 109 cfu/mL) was inoculated, via oral administration at 29 days of age, into ST and HS+ST group birds. On the 4th day after Salmonella Typhimurium administration, an increase in jejunum IgA levels was observed in chickens infected with Salmonella Typhimurium. Mechanistic regulation of TLR4-NFκB-NLRP3 and TLR4-TBK1 signaling by heat stress was evaluated in Salmonella Typhimurium-infected broilers. Heat stress markedly inhibited the expression of cytokines including TNF-α, IL-6, IL-1β, NLRP3, caspase-1, NF-κB-p65, and p-NF-κB-p65, and the TLR4-TBK1 cytokines IFN-α, IFN-γ, p-IRF3, and p-TBK1 in jejunum of broilers infected with Salmonella Typhimurium. Collectively, our results demonstrate that heat stress can inhibit intestinal immune response by downregulating the expression of TLR4-NFκB-NLRP3 and TLR4-TBK1 signaling pathways in broilers infected with Salmonella Typhimurium.
Collapse
Affiliation(s)
- Wei-Hao Li
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Yi-Lei Liu
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Jian-Chi Lun
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Yong-Ming He
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Lu-Ping Tang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
| |
Collapse
|
38
|
Shealy NG, Yoo W, Byndloss MX. Colonization resistance: metabolic warfare as a strategy against pathogenic Enterobacteriaceae. Curr Opin Microbiol 2021; 64:82-90. [PMID: 34688039 DOI: 10.1016/j.mib.2021.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
The intestine is home to a large and complex bacterial ecosystem (microbiota), which performs multiple beneficial functions for the host, including immune education, nutrition, and protection against invasion by enteric pathogens (colonization resistance). The host and microbiome symbiotic interactions occur in part through metabolic crosstalk. Thus, microbiota members have evolved highly diverse metabolic pathways to inhibit pathogen colonization via activation of protective immune responses and nutrient acquisition and utilization. Conversely, pathogenic Enterobacteriaceae actively induce an inflammation-dependent disruption of the gut microbial ecosystem (dysbiosis) to gain a competitive metabolic advantage against the resident microbiota. This review discusses the recent findings on the crucial role of microbiota metabolites in colonization resistance regulation. Additionally, we summarize metabolic mechanisms used by pathogenic Enterobacteriaceae to outcompete commensal microbes and cause disease.
Collapse
Affiliation(s)
- Nicolas G Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Woongjae Yoo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Foley SE, Loew EB, McCormick BA. Recent advances in understanding microbial regulation of host multi-drug resistance transporters. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.100469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
40
|
Inflammasome activation by Salmonella. Curr Opin Microbiol 2021; 64:27-32. [PMID: 34563937 DOI: 10.1016/j.mib.2021.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 11/24/2022]
Abstract
Host recognition of bacteria such as Salmonella enterica serovar Typhimurium requires multiple host detection systems to generate complex inflammatory responses which can be cell type specific and has the potential for bacterial subversion of the host. Host detection of Salmonella requires Pattern Recognition Receptors (PRRs) sensing Pathogen Associated Molecular Patterns (PAMPs). These bacteria possess a diverse array of PAMPs including lipopolysaccharide, flagellin, proteins, lipoproteins, DNA, RNA and metabolites which can potentially activate multiple PRRs concurrently in different cell types. Salmonella is sensed by the inflammasome forming cytosolic nucleotide oligomerisation domain leucine rich repeat-like receptor (NLR) PRRs NLRC4 and NLRP3 as well as by the non canonical inflammasome formed by caspase 11 in mice, caspase 4 and 5 in humans. This review will discuss the different inflammasomes and how their activity regulates the host response to Salmonella infection.
Collapse
|
41
|
Johnson M, Stockdale L, de Haan N, Wuhrer M, Nouta J, Koeleman CAM, Clarke J, Marinou S, Shakya M, Colin-Jones R, Theiss-Nyland K, Voysey M, Jin C, Pant D, Jones E, Kelly S, Dongol S, Karkey A, Shrestha S, Basnyat B, Hill J, Pollard AJ. Association of Antibody-Dependent Neutrophil Phagocytosis With Distinct Antibody Glycosylation Profiles Following Typhoid Vaccination. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.742804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Typhoid Vi-conjugate vaccines (Vi-TCV) have been developed to control typhoid fever in children in endemic regions. Previously, in a human challenge model of typhoid, Vi-TCV was administered prior to deliberate ingestion of Salmonella Typhi by healthy adult volunteers in the UK. Vi-specific antibody-dependent neutrophil phagocytosis (ADNP) was associated with protection against enteric fever in this model, but it is not known if ADNP is induced by vaccination of children. We measured ADNP in a cohort of Nepalese children receiving a Vi-TCV in a field study to investigate whether functional antibody responses were also present in children in an endemic setting. Furthermore, we investigated relationships between the functional antibody measures and other properties of the antibody response, including Vi-IgG and IgA titres, and Fc region glycosylation. Antibody-dependent neutrophil phagocytosis significantly increased in children aged 9 months to 15 years between the day of vaccination and 28 days following administration of Vi-TCV (D28). The magnitude of ADNP was also comparable with the levels of ADNP induced by plasma from vaccinated UK adults. Neither IgG nor IgA antibody titres significantly correlated with ADNP scores at D28; however, increased vaccine-induced ADNP was associated with decreased levels of IgG1 sialylation. These data suggest that vaccination with Vi-TCV produces functional antibody responses in children, which associate with specific glycosylation patterns of the Fc region.
Collapse
|
42
|
Ibarra-Valencia MA, Espino-Solis GP, Estrada BE, Corzo G. Immunomodulatory Responses of Two Synthetic Peptides against Salmonella Typhimurium Infection. Molecules 2021; 26:5573. [PMID: 34577046 PMCID: PMC8466354 DOI: 10.3390/molecules26185573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 12/04/2022] Open
Abstract
In vitro assays of phagocytic activity showed that the peptide Pin2[G] stimulates phagocytosis in BMDM cells from 0.15 to 1.25 μg/mL, and in RAW 264.7 cells at 0.31 μg/mL. In the same way, the peptide FA1 induced phagocytosis in BMDM cells from 1.17 to 4.69 μg/mL and in RAW 264.7 cells at 150 μg/mL. Cytokine profiles of uninfected RAW 264.7 showed that Pin2[G] increased liberation TNF (from 1.25 to 10 μg/mL) and MCP-1 (10 μg/mL), and FA1 also increased the release of TNF (from 18.75 to 75 μg/mL) but did not increase the liberation of MCP-1. In RAW 264.7 macrophages infected with Salmonella enterica serovar Typhimurium, the expression of TNF increases with Pin2[G] (1.25-10 μg/mL) or FA1 (18.75-75 μg/mL). In these cells, FA1 also increases the expression of IL-12p70, IL-10 and IFN-γ when applied at concentrations of 37.5, 75 and 150 μg/mL, respectively. On the other hand, stimulation with 1.25 and 10 μg/mL of Pin2[G] promotes the expression of MCP-1 and IL-12p70, respectively. Finally, peptides treatment did not resolve murine gastric infection, but improves their physical condition. Cytokine profiles showed that FA1 reduces IFN-γ and MCP-1 but increases IL-10, while Pin2[G] reduces IFN-γ but increases the liberation of IL-6 and IL-12p70. This data suggests a promising activity of FA1 and Pin2[G] as immunomodulators of gastric infections in S. Typhimurium.
Collapse
Affiliation(s)
- Marco Antonio Ibarra-Valencia
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, A.P. 510-3, Cuernavaca 62250, Mexico
| | - Gerardo Pável Espino-Solis
- Laboratorio de Investigación Traslacional and Laboratorio Nacional de Citometría de Flujo-UACH, Universidad Autónoma de Chihuahua, Circuito Universitario, Campus II, Chihuahua 31109, Mexico; (G.P.E.-S.); (B.E.E.)
| | - Blanca Elisa Estrada
- Laboratorio de Investigación Traslacional and Laboratorio Nacional de Citometría de Flujo-UACH, Universidad Autónoma de Chihuahua, Circuito Universitario, Campus II, Chihuahua 31109, Mexico; (G.P.E.-S.); (B.E.E.)
| | - Gerardo Corzo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, A.P. 510-3, Cuernavaca 62250, Mexico
| |
Collapse
|
43
|
Buddhasiri S, Sukjoi C, Kaewsakhorn T, Nambunmee K, Nakphaichit M, Nitisinprasert S, Thiennimitr P. Anti-inflammatory Effect of Probiotic Limosilactobacillus reuteri KUB-AC5 Against Salmonella Infection in a Mouse Colitis Model. Front Microbiol 2021; 12:716761. [PMID: 34497597 PMCID: PMC8419263 DOI: 10.3389/fmicb.2021.716761] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/04/2021] [Indexed: 01/31/2023] Open
Abstract
Acute non-typhoidal salmonellosis (NTS) caused by Salmonella enterica Typhimurium (STM) is among the most prevalent of foodborne diseases. A global rising of antibiotic resistance strains of STM raises an urgent need for alternative methods to control this important pathogen. Major human food animals which harbor STM in their gut are cattle, swine, and poultry. Previous studies showed that the probiotic Limosilactobacillus (Lactobacillus) reuteri KUB-AC5 (AC5) exhibited anti-Salmonella activities in chicken by modulating gut microbiota and the immune response. However, the immunobiotic effect of AC5 in a mammalian host is still not known. Here, we investigated the anti-Salmonella and anti-inflammatory effects of AC5 on STM infection using a mouse colitis model. Three groups of C57BL/6 mice (prophylactic, therapeutic, and combined) were fed with 109 colony-forming units (cfu) AC5 daily for 7, 4, and 11 days, respectively. Then, the mice were challenged with STM compared to the untreated group. By using a specific primer pair, we found that AC5 can transiently colonize mouse gut (colon, cecum, and ileum). Interestingly, AC5 reduced STM gut proliferation and invasion together with attenuated gut inflammation and systemic dissemination in mice. The decreased STM numbers in mouse gut lumen, gut tissues, and spleen possibly came from longer AC5 feeding duration and/or the combinatorial (direct and indirect inhibitory) effect of AC5 on STM. However, AC5 attenuated inflammation (both in the gut and in the spleen) with no difference between these three approaches. This study demonstrated that AC5 confers both direct and indirect inhibitory effects on STM in the inflamed gut.
Collapse
Affiliation(s)
- Songphon Buddhasiri
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chutikarn Sukjoi
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thattawan Kaewsakhorn
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kowit Nambunmee
- Major of Occupational Health and Safety, School of Health Science, Mae Fah Luang University, Chiang Rai, Thailand.,Urban Safety Innovation Research Group, Mae Fah Luang University, Chiang Rai, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Parameth Thiennimitr
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, Thailand.,Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
44
|
Stimulation of Toll-Like Receptor 3 Diminishes Intracellular Growth of Salmonella Typhimurium by Enhancing Autophagy in Murine Macrophages. Metabolites 2021; 11:metabo11090602. [PMID: 34564417 PMCID: PMC8466172 DOI: 10.3390/metabo11090602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 12/03/2022] Open
Abstract
The Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative Gram-negative bacterium that causes acute gastroenteritis and food poisoning. S. Typhimurium can survive within macrophages that are able to initiate the innate immune response after recognizing bacteria via various pattern-recognition receptors (PRRs), such as Toll-like receptors (TLRs). In this study, we investigated the effects and molecular mechanisms by which agonists of endosomal TLRs—especially TLR3—contribute to controlling S. Typhimurium infection in murine macrophages. Treatment with polyinosinic:polycytidylic acid (poly(I:C))—an agonist of TLR3—significantly suppressed intracellular bacterial growth by promoting intracellular ROS production in S. Typhimurium-infected cells. Pretreatment with diphenyleneiodonium (DPI)—an NADPH oxidase inhibitor—reduced phosphorylated MEK1/2 levels and restored intracellular bacterial growth in poly(I:C)-treated cells during S. Typhimurium infection. Nitric oxide (NO) production increased through the NF-κB-mediated signaling pathway in poly(I:C)-treated cells during S. Typhimurium infection. Intracellular microtubule-associated protein 1A/1B-light chain 3 (LC3) levels were increased in poly(I:C)-treated cells; however, they were decreased in cells pretreated with 3-methyladenine (3-MA)—a commonly used inhibitor of autophagy. These results suggest that poly(I:C) induces autophagy and enhances ROS production via MEK1/2-mediated signaling to suppress intracellular bacterial growth in S. Typhimurium-infected murine macrophages, and that a TLR3 agonist could be developed as an immune enhancer to protect against S. Typhimurium infection.
Collapse
|
45
|
Abd-Aljabar EM, Jaloob Aljanaby AAJ. ROLE OF MACROPHAGE MIGRATION INHIBITORY FACTOR, CLUSTER OF DIFFERENTIATION 19 AND INTERLEUKIN 23 IN INDIVIDUALS INFECTED WITH Salmonella typhi. JOURNAL OF EXPERIMENTAL BIOLOGY AND AGRICULTURAL SCIENCES 2021; 9:394-400. [DOI: 10.18006/2021.9(3).394.400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Salmonella enterica serovar typhi (S.typhi) is one of the most important gram negative bacteria that cause enteric fever and stimulate the immune response in infected patients. This study aimed to compare the humoral and cellular immune responses in patients infected with S.typhi. For this, the concentration of three immunological markers viz., Macrophage migration inhibitory factor (MIF), Cluster of Differentiation 19 (CD19), and Interleukin 23 (IL-23) were measured by the Enzyme-Linked Immunosorbent Assay in the serum of 54 S.typhi infected patients, and 31 healthy individuals. The results demonstrated a significant increase (P =<0.05) in the serum concentration of MIF in patients infected with acute infection of S. typhi compared with healthy individuals but there were no significant differences in chronic infection. While there was a significant increase (P =<0.05) in serum concentration of CD19 and IL-23 in patients infected with acute and chronic infection of S.typhi as compared with healthy individuals. In conclusion, macrophage migration inhibitory factor plays a vital role in patients infected with acute infection of S. typhi and has no synergism with T-cell and B-cell in acute and chronic infection.
Collapse
|
46
|
Agbayani G, Clark K, Sandhu JK, Hewitt M, Sad S, Murphy SP, Krishnan L. IFN-alpha receptor deficiency enhances host resistance to oral Salmonella enterica serovar Typhimurium infection during murine pregnancy. Am J Reprod Immunol 2021; 86:e13454. [PMID: 33991140 DOI: 10.1111/aji.13454] [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] [Received: 03/01/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
PROBLEM Maternal tolerance during pregnancy increases the risk of infection with certain intracellular pathogens. Systemic Salmonella enterica serovar Typhimurium (S.Tm) infection during pregnancy in normally resistant 129X1/SvJ mice leads to severe placental infection, as well as fetal and maternal deaths. However, the effect of oral infection with S.Tm in pregnant mice and the roles of infection-induced inflammation and cell death pathways in contributing to susceptibility to infection are unclear. METHOD OF STUDY Non-pregnant and pregnant C57BL/6J wild-type (WT) and cell death pathway-altered mice (IFNAR1-/- , Caspase-1, 11-/- , RIP3-/- ) were infected orally with S.Tm. Host survival and fetal resorption were determined. Bacterial burden in mesenteric lymph nodes (MLNs), spleen, liver, and placentas was enumerated at various time points post-infection. Serum cytokine expression was measured through cytometric bead array. RESULTS Oral infection of WT mice with S.Tm on days 9-10 of gestation resulted in systemic dissemination of the bacteria, substantial placental colonization, and fetal loss 5 days post-infection. Histopathological examination of the placentas indicated that infection-induced widespread focal necrosis and neutrophil infiltration throughout the spongiotrophoblast (SpT) layer. In the non-pregnant state, IFNAR1-/- mice exhibited increased survival following oral S.Tm infection relative to Caspase-1, 11-/- , RIP3-/- , and WT mice. The increased resistance to S.Tm infection in IFNAR1-/- mice was seen during pregnancy as well, with decreased bacterial burden within MLNs, spleen, and placenta, which correlated with the decreased resorptions relative to WT and Caspase-1, 11-/- mice. CONCLUSION Oral S.Tm exposure leads to placental infection, inflammation, and resorption, whereas IFNAR1 deficiency enhances host resistance both in the non-pregnant and pregnant states.
Collapse
Affiliation(s)
- Gerard Agbayani
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Kristina Clark
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Shawn P Murphy
- Department of Obstetrics and Gynecology, University of Rochester, Rochester, NY, USA.,Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Lakshmi Krishnan
- Division of Life Sciences, Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
47
|
Erazo AB, Wang N, Standke L, Semeniuk AD, Fülle L, Cengiz SC, Thiem M, Weighardt H, Strugnell RA, Förster I. CCL17-expressing dendritic cells in the intestine are preferentially infected by Salmonella but CCL17 plays a redundant role in systemic dissemination. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:891-904. [PMID: 33945673 PMCID: PMC8342217 DOI: 10.1002/iid3.445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Introduction Salmonella spp. are a recognized and global cause of serious health issues from gastroenteritis to invasive disease. The mouse model of human typhoid fever, which uses Salmonella enterica serovar Typhimurium (STM) in susceptible mouse strains, has revealed that the bacteria gain access to extraintestinal tissues from the gastrointestinal tract to cause severe systemic disease. Previous analysis of the immune responses against Salmonella spp. revealed the crucial role played by dendritic cells (DCs) in carrying STM from the intestinal mucosa to the mesenteric lymph nodes (mLNs), a key site for antigen presentation and T cell activation. In this study, we investigated the influence of chemokine CCL17 on the dissemination of STM. Methods WT, CCL17/EGFP reporter, or CCL17‐deficient mice were infected orally with STM (SL1344) or mCherry‐expressing STM for 1–3 days. Colocalization of STM with CCL17‐expressing DCs in Peyer's patches (PP) and mLN was analyzed by fluorescence microscopy. In addition, DCs and myeloid cell populations from naïve and Salmonella‐infected mice were analyzed by flow cytometry. Bacterial load was determined in PP, mLN, spleen, and liver 1 and 3 days after infection. Results Histological analysis revealed that CCL17‐expressing cells are located in close proximity to STM in the dome area of PP. We show that, in mLN, STM were preferentially located within CCL17+ rather than CCL17− DCs, besides other mononuclear phagocytes, and identified the CD103+ CD11b− DC subset as the main STM‐carrying DC population in the intestine. STM infection triggered upregulation of CCL17 expression in specific intestinal DC subsets in a tissue‐specific manner. The dissemination of STM from the gut to the mLN, however, was only moderately influenced by the presence of CCL17. Conclusion CCL17‐expressing DCs were preferentially infected by Salmonella in the intestine in comparison to other DC. Nevertheless, the production of CCL17 was not essential for the early dissemination of Salmonella from the gut to systemic organs.
Collapse
Affiliation(s)
- Anna B Erazo
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lena Standke
- Department for Innate Immunity and Metaflammation, Institute of Innate Immunity, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Adrian D Semeniuk
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lorenz Fülle
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sevgi C Cengiz
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manja Thiem
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Heike Weighardt
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Richard A Strugnell
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
48
|
Pritsch M, Ben Khaled N, Liegl G, Schubert S, Hoelscher M, Woischke C, Arens N, Thorn-Seshold J, Kammermeier S, Wieser A. Rapid prototyping vaccine approach in mice against multi-drug resistant Gram-negative organisms from clinical isolates based on outer membrane vesicles. Microbiol Immunol 2021; 65:214-227. [PMID: 33650163 DOI: 10.1111/1348-0421.12882] [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] [Received: 10/17/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Hospital-acquired infections due to multi-drug resistant Gram-negative organisms (MDRGNO) pose a major threat to global health. A vaccine preventing colonization and consecutive infection with MDRGNO could be particularly valuable, as therapeutic options become increasingly limited. Outer membrane vesicles (OMV) of Escherichia coli strain CFT073 as well as three MDRGNO strains that had caused severe infections in humans were administered intranasally to mice, with and without cholera toxin as an adjuvant. The humoral immune responses were comparatively matched with the sera of patients, who had suffered an infection caused by the respective bacterium. Additionally, systemic and local toxicity was evaluated. Intranasal vaccination with OMV could elicit solid humoral immune responses (total IgM and IgG), specific for the respective MDRGNO in mice; decoration of vital bacterial membranes with antibodies was comparable to patients who had survived systemic infection with the respective bacterial isolate. After intranasal vaccination of mice with OMV no signs of local or systemic toxicity were observed. Intranasal vaccination with OMV may open up a rapid vaccine approach to prevent colonization and/or infection with pathogenic MDRGNOs, especially in an outbreak setting within a hospital. It may also be an option for patients who have to undergo elective interventions in centers with a high risk of infection for certain common MDRGNO. Future studies need to include challenge experiments as well as phase I trials in humans.
Collapse
Affiliation(s)
- Michael Pritsch
- Department of Bacteriology, Max von Pettenkofer-Institute (LMU), Munich, Germany.,Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,3CIHLMU Center for International Health, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Najib Ben Khaled
- Department of Bacteriology, Max von Pettenkofer-Institute (LMU), Munich, Germany.,Department of Medicine II, University Hospital Munich (LMU), Munich, Germany
| | - Gabriele Liegl
- Department of Bacteriology, Max von Pettenkofer-Institute (LMU), Munich, Germany
| | - Soeren Schubert
- Department of Bacteriology, Max von Pettenkofer-Institute (LMU), Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,3CIHLMU Center for International Health, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Christine Woischke
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Nathalie Arens
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,Department of Medicine IV, University Hospital Augsburg, Augsburg, Germany
| | | | - Stefan Kammermeier
- Department of Neurology, University Hospital Munich (LMU), Munich, Germany
| | - Andreas Wieser
- Department of Bacteriology, Max von Pettenkofer-Institute (LMU), Munich, Germany.,Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany.,3CIHLMU Center for International Health, LMU Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| |
Collapse
|
49
|
Transcriptome Analysis Reveals Possible Immunomodulatory Activity Mechanism of Chlorella sp. Exopolysaccharides on RAW264.7 Macrophages. Mar Drugs 2021; 19:md19040217. [PMID: 33919822 PMCID: PMC8070752 DOI: 10.3390/md19040217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
In this study, the exopolysaccharides of Chlorella sp. (CEP) were isolated to obtain the purified fraction CEP4. Characterization results showed that CEP4 was a sulfated heteropolysaccharide. The main monosaccharide components of CEP4 are glucosamine hydrochloride (40.8%) and glucuronic acid (21.0%). The impact of CEP4 on the immune activity of RAW264.7 macrophage cytokines was detected, and the results showed that CEP4 induced the production of nitric oxide (NO), TNF-α, and IL-6 in a dose-dependent pattern within a range of 6 μg/mL. A total of 4824 differentially expressed genes (DEGs) were obtained from the results of RNA-seq. Gene enrichment analysis showed that immune-related genes such as NFKB1, IL-6, and IL-1β were significantly upregulated, while the genes RIPK1 and TLR4 were significantly downregulated. KEGG pathway enrichment analysis showed that DEGs were significantly enriched in immune-related biological processes, including toll-like receptor (TLR) signaling pathway, cytosolic DNA-sensing pathway, and C-type lectin receptor signaling pathway. Protein–protein interaction (PPI) network analysis showed that HSP90AB1, Rbx1, ISG15, Psmb6, Psmb3, Psmb8, PSMA7, Polr2f, Rpsa, and NEDD8 were the hub genes with an essential role in the immune activity of CEP4. The preliminary results of the present study revealed the potential mechanism of CEP4 in the immune regulation of RAW264.7 macrophages, suggesting that CEP4 is a promising immunoregulatory agent.
Collapse
|
50
|
Evaluating the effect of spaceflight on the host-pathogen interaction between human intestinal epithelial cells and Salmonella Typhimurium. NPJ Microgravity 2021; 7:9. [PMID: 33750813 PMCID: PMC7943786 DOI: 10.1038/s41526-021-00136-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Spaceflight uniquely alters the physiology of both human cells and microbial pathogens, stimulating cellular and molecular changes directly relevant to infectious disease. However, the influence of this environment on host-pathogen interactions remains poorly understood. Here we report our results from the STL-IMMUNE study flown aboard Space Shuttle mission STS-131, which investigated multi-omic responses (transcriptomic, proteomic) of human intestinal epithelial cells to infection with Salmonella Typhimurium when both host and pathogen were simultaneously exposed to spaceflight. To our knowledge, this was the first in-flight infection and dual RNA-seq analysis using human cells.
Collapse
|