1
|
Dong Y, Meng F, Wang J, Wei J, Zhang K, Qin S, Li M, Wang F, Wang B, Liu T, Zhong W, Cao H. Desulfovibrio vulgaris flagellin exacerbates colorectal cancer through activating LRRC19/TRAF6/TAK1 pathway. Gut Microbes 2025; 17:2446376. [PMID: 39718561 DOI: 10.1080/19490976.2024.2446376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/22/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
The initiation and progression of colorectal cancer (CRC) are intimately associated with genetic, environmental and biological factors. Desulfovibrio vulgaris (DSV), a sulfate-reducing bacterium, has been found excessive growth in CRC patients, suggesting a potential role in carcinogenesis. However, the precise mechanisms underlying this association remain incompletely understood. We have found Desulfovibrio was abundant in high-fat diet-induced Apcmin/+ mice, and DSV, a member of Desulfovibrio, triggered colonocyte proliferation of germ-free mice. Furthermore, the level of DSV progressively rose from healthy individuals to CRC patients. Flagella are important accessory structures of bacteria, which can help them colonize and enhance their invasive ability. We found that D. vulgaris flagellin (DVF) drove the proliferation, migration, and invasion of CRC cells and fostered the growth of CRC xenografts. DVF enriched the epithelial-mesenchymal transition (EMT)-associated genes and characterized the facilitation of DVF on EMT. Mechanistically, DVF induced EMT through a functional transmembrane receptor called leucine-rich repeat containing 19 (LRRC19). DVF interacted with LRRC19 to modulate the ubiquitination of tumor necrosis factor receptor-associated factor (TRAF)6, rather than TRAF2. This interaction drove the ubiquitination of pivotal molecule TAK1, further enhancing its autophosphorylation and ultimately contributing to EMT. Collectively, DVF interacts with LRRC19 to activate the TRAF6/TAK1 signaling pathway, thereby promoting the EMT of CRC. These data shed new light on the role of gut microbiota in CRC and establish a potential clinical therapeutic target.
Collapse
Affiliation(s)
- Yue Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fanyi Meng
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingyi Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kexin Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Siqi Qin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fucheng Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
2
|
Afzal H, Murtaza A, Cheng LT. Structural engineering of flagellin as vaccine adjuvant: quest for the minimal domain of flagellin for TLR5 activation. Mol Biol Rep 2025; 52:104. [PMID: 39775323 PMCID: PMC11706886 DOI: 10.1007/s11033-024-10146-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Flagellin stimulates Toll-like receptor 5 (TLR5), triggering both innate and adaptive immune responses, making it a potential vaccine adjuvant. On mucosal surfaces, flagellin induces a strong release of cytokines, chemokines, and immunoglobulins. When used in its free monomeric form, flagellin has been shown to enhance immune responses when combined with vaccine antigens. Further research demonstrated that genetically linking flagellin to the antigen provides a more consistent immune boost. However, the bulky structure of flagellin presents challenges in designing the antigen-adjuvant construct, leading to ongoing research to determine the minimal flagellin domain necessary for its adjuvant effect. Early findings suggest that only the D0 and D1 domains are required for immune enhancement. Functional analysis revealed that the TLR5-binding region is located in the D1 domain, while TLR5 dimerization and signaling require the presence of D0. Further reductions in the size of the D0 and D1 domains may be possible as deeper studies aim to identify the key residues responsible for TLR5 activation and immune enhancement. Additionally, flagellin is being tested as a hapten carrier alongside its established adjuvant role. Recently, significant advancements in flagellin application have been observed as it progresses through clinical studies as an adjuvant, anti-radiation, and anti-cancer agent.
Collapse
Affiliation(s)
- Haroon Afzal
- International Degree Program of Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan
| | - Asad Murtaza
- International Degree Program of Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan.
- Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, Tromsø, Norway.
| | - Li-Ting Cheng
- International Degree Program of Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan.
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan.
| |
Collapse
|
3
|
Omole Z, Dorrell N, Elmi A, Nasher F, Gundogdu O, Wren BW. Pathogenicity and virulence of Campylobacter jejuni: What do we really know? Virulence 2024; 15:2436060. [PMID: 39648291 PMCID: PMC11633169 DOI: 10.1080/21505594.2024.2436060] [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: 06/14/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 12/10/2024] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial gastroenteritis and is a major public health concern worldwide. Despite its importance, our understanding of how C. jejuni causes diarrhoea and interacts with its hosts is limited due to the absence of appropriate infection models and established virulence factors found in other enteric pathogens. Additionally, despite its genetic diversity, non-pathogenic C. jejuni strains are unknown. Regardless of these limitations, significant progress has been made in understanding how C. jejuni uses a complex array of factors which aid the bacterium to survive and respond to host defences. This review provides an update on fitness and virulence determinants of this important pathogen and questions our knowledge on these determinants that are often based on inferred genomics knowledge and surrogate infection models.
Collapse
Affiliation(s)
- Zahra Omole
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Nick Dorrell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Abdi Elmi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Fauzy Nasher
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Brendan W. Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
4
|
Cui J, Batley KC, Silver LW, McLennan EA, Hogg CJ, Belov K. Spatial variation in toll-like receptor diversity in koala populations across their geographic distribution. Immunogenetics 2024; 77:5. [PMID: 39614880 PMCID: PMC11608166 DOI: 10.1007/s00251-024-01365-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 01/29/2025]
Abstract
The koala (Phascolarctos cinereus) is an iconic Australian species that is listed as endangered in the northern parts of its range due to loss of habitat, disease, and road deaths. Diseases contribute significantly to the decline of koala populations, primarily Chlamydia and koala retrovirus. The distribution of these diseases across the species' range, however, is not even. Toll-like receptors (TLRs) play a crucial role in innate immunity by recognising and responding to various pathogens. Variations in TLR genes can influence an individual's susceptibility or resistance to infectious diseases. The aim of this study was to identify koala TLR diversity across the east coast of Australia using 413 re-sequenced genomes at 30 × coverage. We identified 45 single-nucleotide polymorphisms (SNP) leading to 51 alleles within ten TLR genes. Our results show that the diversity of TLR genes in the koala forms four distinct genetic groups, which are consistent with the diversity of the koala major histocompatibility complex (MHC), another key immune gene family. The bioinformatics approach presented here has broad applicability to other threatened species with existing genomic resources.
Collapse
Affiliation(s)
- Jian Cui
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kimberley C Batley
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Luke W Silver
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, Australia
| | - Elspeth A McLennan
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Carolyn J Hogg
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, Australia.
| | - Katherine Belov
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Pang S, Liu M, Wang L, Shao M, Zhu G, Duan Q. Differential Adjuvant Activity by Flagellins from Escherichia coli, Salmonella enterica Serotype Typhimurium, and Pseudomonas aeruginosa. Vaccines (Basel) 2024; 12:1212. [PMID: 39591115 PMCID: PMC11598095 DOI: 10.3390/vaccines12111212] [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: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
(1) Background: The adjuvant properties of flagellin from various bacterial species have been extensively studied; however, a systematic comparison of the immunoadjuvant effects of flagellins from different bacterial species is lacking. This study aims to analyze the amino acid sequences and structural features of flagellins from Escherichia coli (FliCE.C), Salmonella enterica serotype Typhimurium (FliCS.T), and Pseudomonas aeruginosa (FliCP.A), and to evaluate their adjuvant activities in terms of Toll-like receptor 5 (TLR5) activation, antibody production, and cytokine responses in a murine model. (2) Methods: Bioinformatics analysis was conducted to compare the amino acid sequences and structural domains (D0, D1, D2, and D3) of flagellins from the three bacterial species. PyMol atomic models were used to confirm structural differences. Toll-like receptor 5 (TLR5) activation assays were performed to measure IL-8 and TNF-α production in vitro. The IgG antibody titers against the model antigen FaeG and cytokine responses, including IL-4 and TNF-α secretion were evaluated in a murine model. (3) Results: Bioinformatics analysis revealed that the D0 and D1 domains are highly conserved, whereas the D2 and D3 domains exhibit significant variability across the three species. Structural analysis via PyMol confirmed these differences, particularly in the D2 and D3 domains. TLR5 activation assays showed that FliCS.T and FliCP.A induced higher levels of IL-8 and TNF-α production compared to FliCE.C, indicating species-specific variations in TLR5 activation. In the murine model, FliCS.T as an adjuvant produced higher antibody titers against FaeG and increased IL-4 secretion in splenocytes compared to FliCE.C and FliCP.A. FliCP.A induced higher TNF-α expression than FliCS.T and FliCE.C, suggesting FliCS.T and FliCP.A are more effective at inducing T-cell responses. (4) Conclusions: This study highlights the potential of FliCS.T and FliCP.A as potent vaccine adjuvants. The results provide insights into the structure-function relationships of these flagellins and support their application in enhancing immune responses against diverse pathogens.
Collapse
Affiliation(s)
- Shengmei Pang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Mei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Longlong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Mingqing Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (S.P.); (M.L.); (L.W.); (M.S.); (G.Z.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
6
|
Eser E, Felton VA, Drolia R, Bhunia AK. Salmonella Detection in Food Using a HEK-hTLR5 Reporter Cell-Based Sensor. BIOSENSORS 2024; 14:444. [PMID: 39329819 PMCID: PMC11430776 DOI: 10.3390/bios14090444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/31/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024]
Abstract
The development of a rapid, sensitive, specific method for detecting foodborne pathogens is paramount for supplying safe food to enhance public health safety. Despite the significant improvement in pathogen detection methods, key issues are still associated with rapid methods, such as distinguishing living cells from dead, the pathogenic potential or health risk of the analyte at the time of consumption, the detection limit, and the sample-to-result. Mammalian cell-based assays analyze pathogens' interaction with host cells and are responsive only to live pathogens or active toxins. In this study, a human embryonic kidney (HEK293) cell line expressing Toll-Like Receptor 5 (TLR-5) and chromogenic reporter system (HEK dual hTLR5) was used for the detection of viable Salmonella in a 96-well tissue culture plate. This cell line responds to low concentrations of TLR5 agonist flagellin. Stimulation of TLR5 ligand activates nuclear factor-kB (NF-κB)-linked alkaline phosphatase (AP-1) signaling cascade inducing the production of secreted embryonic alkaline phosphatase (SEAP). With the addition of a ρ-nitrophenyl phosphate as a substrate, a colored end product representing a positive signal is quantified. The assay's specificity was validated with the top 20 Salmonella enterica serovars and 19 non-Salmonella spp. The performance of the assay was also validated with spiked food samples. The total detection time (sample-to-result), including shortened pre-enrichment (4 h) and selective enrichment (4 h) steps with artificially inoculated outbreak-implicated food samples (chicken, peanut kernel, peanut butter, black pepper, mayonnaise, and peach), was 15 h when inoculated at 1-100 CFU/25 g sample. These results show the potential of HEK-DualTM hTLR5 cell-based functional biosensors for the rapid screening of Salmonella.
Collapse
Affiliation(s)
- Esma Eser
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Department of Food Engineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Victoria A Felton
- Molecular and Cellular Microbiology Laboratory, Department of Biological Science, Old Dominion University, Norfolk, VA 23529, USA
| | - Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Molecular and Cellular Microbiology Laboratory, Department of Biological Science, Old Dominion University, Norfolk, VA 23529, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Center for Bioelectronics, Old Dominion University, Norfolk, VA 23508, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
7
|
Li S, Wen M, Wen G, Yang Y. Structure and biological activity in vitro of Flagellin and its mutants from Escherichia coli Nissle 1917. Arch Microbiol 2024; 206:221. [PMID: 38637410 DOI: 10.1007/s00203-024-03907-7] [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: 01/18/2024] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 04/20/2024]
Abstract
Bacterial flagellin is a potent immunomodulatory agent. Previously, we successfully obtained flagellin from Escherichia coli Nissle 1917 (FliCEcN) and constructed two mutants with varying degrees of deletion in its highly variable regions (HVRs). We found that there was a difference in immune stimulation levels between the two mutants, with the mutant lacking the D2-D3 domain pair of FliCEcN having a better adjuvant effect. Therefore, this study further analyzed the structural characteristics of the aforementioned FliCEcN and its two mutants and measured their levels of Caco-2 cell stimulation to explore the impact of different domains in the HVRs of FliCEcN on its structure and immune efficacy. This study utilized AlphaFold2, SERS (Surface-enhanced Raman spectroscopy), and CD (circular dichroism) techniques to analyze the structural characteristics of FliCEcN and its mutants, FliCΔ174-506 and FliCΔ274-406, and tested their immune effects by stimulating Caco-2 cells in vitro. The results indicate that the D2 and D3 domains of FliCEcN have more complex interactions compared to the D1-D2 domain pair., and these domains also play a role in molecular docking with TLR5 (Toll-like receptor 5). Furthermore, FliCΔ274-406 has more missing side chain and characteristic amino acid peaks than FliCΔ174-506. The FliCEcN group was found to stimulate higher levels of IL-10 (interleukin 10) secretion, while the FliCΔ174-506 and FliCΔ274-406 groups had higher levels of IL-6 (interleukin 6) and TNF-α (tumor necrosis factor-α) secretion. In summary, the deletion of different domains in the HVRs of FliCEcN affects its structural characteristics, its interaction with TLR5, and the secretion of immune factors by Caco-2 cells.
Collapse
Affiliation(s)
- Shuang Li
- Faculty of Animal Science, Department of Veterinary Medicine, Guizhou University, Guiyang, 550025, China
| | - Ming Wen
- Faculty of Animal Science, Department of Veterinary Medicine, Guizhou University, Guiyang, 550025, China
| | - Guilan Wen
- Faculty of Animal Science, Department of Veterinary Medicine, Guizhou University, Guiyang, 550025, China
| | - Ying Yang
- Faculty of Animal Science, Department of Veterinary Medicine, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
8
|
Liao Z, Ji W, Yang C, Su J. TLR5M and TLR5S Synergistically Sense Flagellin in Early Endosome in Lamprey Petromyzon marinus, Switched by the N-Glycosylation Site N239. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:855-867. [PMID: 38231121 DOI: 10.4049/jimmunol.2300490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
In mammals, TLR5 functions as a homodimer to recognize bacterial flagellin on the cytomembrane. The current investigations reveal the existence of two types of TLR5, a membrane-bound PmTLR5M, and a soluble variant PmTLR5S, in lamprey (Petromyzon marinus). Although both PmTLR5M and PmTLR5S can bind flagellin, only PmTLR5M is capable of eliciting a proinflammatory response, whereas PmTLR5S can detect the flagellin and facilitate the role of PmTLR5M in early endosomes. The trafficking chaperone UNC93B1 enhances the ligand-induced signaling via PmTLR5M or the combination of PmTLR5M and PmTLR5S. PmTLR5M recruits MyD88 as an adaptor. Furthermore, chimeric receptor studies demonstrate the indispensability of the intradomain of PmTLR5M in effective activation of the proinflammatory pathway upon flagellin stimulation, and the combination of PmTLR5S with a singular intradomain in both homodimer and heterodimer ectodomain arrangements can very significantly augment the immune response. Furthermore, the flagellin binding sites between PmTLR5M and PmTLR5S are conserved, which are essential for ligand binding and signal transduction. Moreover, investigations on N-linked glycosylation modifications reveal that the N239 site in PmTLR5M and PmTLR5S plays a switch role in both flagellin binding and immune responses. In addition, PmTLR5M exhibits the high-mannose-type and complex-type N-glycosylation modifications; however, PmTLR5S shows exclusive complex-type N-glycosylation modification. The key N239 site demonstrates complex-type N-glycosylation modification. The findings address the function and mechanism of TLR5 in ligand recognition, subcellular localization, and signaling pathway in lowest vertebrate and immune system transition species, highlight the regulatory role of N-glycosylation modification in TLRs, and augment immune evolutionary research on the TLR signaling pathway.
Collapse
Affiliation(s)
- Zhiwei Liao
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wei Ji
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
9
|
Rezayatmand H, Golestani N, Haghighat Hoseini AS, Mousavialmaleki E, Alem M, Farzane Yegane D. Gene expression profile of Campylobacter jejuni in response to macrolide antibiotics. Arch Microbiol 2024; 206:117. [PMID: 38393387 DOI: 10.1007/s00203-024-03849-0] [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: 11/18/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024]
Abstract
Campylobacter jejuni is a foodborne pathogen that causes gastroenteritis in humans and has developed resistance to various antibiotics. The primary objective of this research was to examine the network of antibiotic resistance in C. jejuni. The study involved the wild and antibiotic-resistant strains placed in the presence and absence of antibiotics to review their gene expression profiles in response to ciprofloxacin via microarray. Differentially expressed genes (DEGs) analysis and Protein-Protein Interaction (PPI) Network studies were performed for these genes. The results showed that the resistance network of C. jejuni is modular, with different genes involved in bacterial motility, capsule synthesis, efflux, and amino acid and sugar synthesis. Antibiotic treatment resulted in the down-regulation of cluster genes related to translation, flagellum formation, and chemotaxis. In contrast, cluster genes involved in homeostasis, capsule formation, and cation efflux were up-regulated. The study also found that macrolide antibiotics inhibit the progression of C. jejuni infection by inactivating topoisomerase enzymes and increasing the activity of epimerase enzymes, trying to compensate for the effect of DNA twisting. Then, the bacterium limits the movement to conserve energy. Identifying the antibiotic resistance network in C. jejuni can aid in developing drugs to combat these bacteria. Genes involved in cell division, capsule formation, and substance transport may be potential targets for inhibitory drugs. Future research must be directed toward comprehending the underlying mechanisms contributing to the modularity of antibiotic resistance and developing strategies to disrupt and mitigate the growing threat of antibiotic resistance effectively.
Collapse
Affiliation(s)
- Hamed Rezayatmand
- Department of Pharmacy and Technology of Organic Substances, Industrial Pharmacy, Ukrainian State Chemical Technology University, Dnipro, Ukraine
| | - Nafiseh Golestani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Elaheh Mousavialmaleki
- Pharmaceutical Science Research Center, Medicinal Chemistry Department, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahsa Alem
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Donya Farzane Yegane
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
10
|
Zhao H, Xu Y, Yang L, Wang Y, Li M, Chen L. Biological Function of Prophage-Related Gene Cluster Δ VpaChn25_RS25055~Δ VpaChn25_0714 of Vibrio parahaemolyticus CHN25. Int J Mol Sci 2024; 25:1393. [PMID: 38338671 PMCID: PMC10855970 DOI: 10.3390/ijms25031393] [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: 12/14/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024] Open
Abstract
Vibrio parahaemolyticus is the primary foodborne pathogen known to cause gastrointestinal infections in humans. Nevertheless, the molecular mechanisms of V. parahaemolyticus pathogenicity are not fully understood. Prophages carry virulence and antibiotic resistance genes commonly found in Vibrio populations, and they facilitate the spread of virulence and the emergence of pathogenic Vibrio strains. In this study, we characterized three such genes, VpaChn25_0713, VpaChn25_0714, and VpaChn25_RS25055, within the largest prophage gene cluster in V. parahaemolyticus CHN25. The deletion mutants ΔVpaChn25_RS25055, ΔVpaChn25_0713, ΔVpaChn25_0714, and ΔVpaChn25_RS25055-0713-0714 were derived with homologous recombination, and the complementary mutants ΔVpaChn25_0713-com, ΔVpaChn25_0714-com, ΔVpaChn25_RS25055-com, ΔVpaChn25_RS25055-0713-0714-com were also constructed. In the absence of the VpaChn25_RS25055, VpaChn25_0713, VpaChn25_0714, and VpaChn25_RS25055-0713-0714 genes, the mutants showed significant reductions in low-temperature survivability and biofilm formation (p < 0.001). The ΔVpaChn25_0713, ΔVpaChn25_RS25055, and ΔVpaChn25_RS25055-0713-0714 mutants were also significantly defective in swimming motility (p < 0.001). In the Caco-2 model, the above four mutants attenuated the cytotoxic effects of V. parahaemolyticus CHN25 on human intestinal epithelial cells (p < 0.01), especially the ΔVpaChn25_RS25055 and ΔVpaChn25_RS25055-0713-0714 mutants. Transcriptomic analysis showed that 15, 14, 8, and 11 metabolic pathways were changed in the ΔVpaChn25_RS25055, ΔVpaChn25_0713, ΔVpaChn25_0714, and ΔVpaChn25_RS25055-0713-0714 mutants, respectively. We labeled the VpaChn25_RS25055 gene with superfolder green fluorescent protein (sfGFP) and found it localized at both poles of the bacteria cell. In addition, we analyzed the evolutionary origins of the above genes. In summary, the prophage genes VpaChn25_0713, VpaChn25_0714, and VpaChn25_RS25055 enhance V. parahaemolyticus CHN25's survival in the environment and host. Our work improves the comprehension of the synergy between prophage-associated genes and the evolutionary process of V. parahaemolyticus.
Collapse
Affiliation(s)
- Hui Zhao
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (H.Z.); (Y.X.); (L.Y.)
| | - Yingwei Xu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (H.Z.); (Y.X.); (L.Y.)
| | - Lianzhi Yang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (H.Z.); (Y.X.); (L.Y.)
| | - Yaping Wang
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Centre, Richmond, VA 23284, USA;
| | - Mingyou Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
| | - Lanming Chen
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (H.Z.); (Y.X.); (L.Y.)
| |
Collapse
|
11
|
Lim JS, Jeon EJ, Go HS, Kim HJ, Kim KY, Nguyen TQT, Lee DY, Kim KS, Pietrocola F, Hong SH, Lee SE, Kim KS, Park TS, Choi DH, Jeong YJ, Park JH, Kim HS, Min JJ, Kim YS, Park JT, Cho JH, Lee GW, Lee JH, Choy HE, Park SC, Lee CH, Rhee JH, Serrano M, Cho KA. Mucosal TLR5 activation controls healthspan and longevity. Nat Commun 2024; 15:46. [PMID: 38167804 PMCID: PMC10761998 DOI: 10.1038/s41467-023-44263-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Addressing age-related immunological defects through therapeutic interventions is essential for healthy aging, as the immune system plays a crucial role in controlling infections, malignancies, and in supporting tissue homeostasis and repair. In our study, we show that stimulating toll-like receptor 5 (TLR5) via mucosal delivery of a flagellin-containing fusion protein effectively extends the lifespan and enhances the healthspan of mice of both sexes. This enhancement in healthspan is evidenced by diminished hair loss and ocular lens opacity, increased bone mineral density, improved stem cell activity, delayed thymic involution, heightened cognitive capacity, and the prevention of pulmonary lung fibrosis. Additionally, this fusion protein boosts intestinal mucosal integrity by augmenting the surface expression of TLR5 in a certain subset of dendritic cells and increasing interleukin-22 (IL-22) secretion. In this work, we present observations that underscore the benefits of TLR5-dependent stimulation in the mucosal compartment, suggesting a viable strategy for enhancing longevity and healthspan.
Collapse
Affiliation(s)
- Jae Sung Lim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Eun Jae Jeon
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, 13486, Republic of Korea
| | - Hye Sun Go
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, 13486, Republic of Korea
| | - Hyung-Jin Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, 13486, Republic of Korea
| | - Kye Young Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, 13486, Republic of Korea
| | - Thi Quynh Trang Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Da Young Lee
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Kyu Suk Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Federico Pietrocola
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Seol Hee Hong
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Shee Eun Lee
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Tae-Shin Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dong-Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Yu-Jin Jeong
- Department of Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jong-Hwan Park
- Department of Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyeon Sik Kim
- Medical Photonic Research Center, Korea Photonics Technology Institute, Gwangju, 61007, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Joon Tae Park
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Jae-Ho Cho
- Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Gil-Woo Lee
- Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Ji Hyeon Lee
- Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Sang Chul Park
- Future Life and Society Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
| | - Joon Haeng Rhee
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
- Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
- Department of Microbiology, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| | - Kyung A Cho
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, 13486, Republic of Korea.
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
- Future Life and Society Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
| |
Collapse
|
12
|
Temchura V, Wagner JT, Damm D. Immunogenicity of Recombinant Lipid-Based Nanoparticle Vaccines: Danger Signal vs. Helping Hand. Pharmaceutics 2023; 16:24. [PMID: 38258035 PMCID: PMC10818441 DOI: 10.3390/pharmaceutics16010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Infectious diseases are a predominant problem in human health. While the incidence of many pathogenic infections is controlled by vaccines, some pathogens still pose a challenging task for vaccine researchers. In order to face these challenges, the field of vaccine development has changed tremendously over the last few years. For non-replicating recombinant antigens, novel vaccine delivery systems that attempt to increase the immunogenicity by mimicking structural properties of pathogens are already approved for clinical applications. Lipid-based nanoparticles (LbNPs) of different natures are vesicles made of lipid layers with aqueous cavities, which may carry antigens and other biomolecules either displayed on the surface or encapsulated in the cavity. However, the efficacy profile of recombinant LbNP vaccines is not as high as that of live-attenuated ones. This review gives a compendious picture of two approaches that affect the immunogenicity of recombinant LbNP vaccines: (i) the incorporation of immunostimulatory agents and (ii) the utilization of pre-existing or promiscuous cellular immunity, which might be beneficial for the development of tailored prophylactic and therapeutic LbNP vaccine candidates.
Collapse
Affiliation(s)
- Vladimir Temchura
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | | | - Dominik Damm
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
13
|
Pei C, Dong H, Teng Z, Wei S, Zhang Y, Yin S, Tang J, Sun S, Guo H. Self-Assembling Nanovaccine Fused with Flagellin Enhances Protective Effect against Foot-and-Mouth Disease Virus. Vaccines (Basel) 2023; 11:1675. [PMID: 38006007 PMCID: PMC10675102 DOI: 10.3390/vaccines11111675] [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: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Nanovaccines based on self-assembling nanoparticles (NPs) can show conformational epitopes of antigens and they have high immunogenicity. In addition, flagellin, as a biological immune enhancer, can be fused with an antigen to considerably enhance the immune effect of antigens. In improving the immunogenicity and stability of a foot-and-mouth disease virus (FMDV) antigen, novel FMDV NP antigens were prepared by covalently coupling the VP1 protein and truncated flagellin containing only N-terminus D0 and D1 (N-terminal aa 1-99, nFLiC) with self-assembling NPs (i301). The results showed that the fusion proteins VP1-i301 and VP1-i301-nFLiC can assemble into NPs with high thermal tolerance and stability, obtain high cell uptake efficiency, and upregulate marker molecules and immune-stimulating cytokines in vitro. In addition, compared with monomeric VP1 antigen, high-level cytokines were stimulated with VP1-i301 and VP1-i301-nFLiC nanovaccines in guinea pigs, to provide clinical protection against viral infection comparable to an inactivated vaccine. This study provides new insight for the development of a novel FMD vaccine.
Collapse
Affiliation(s)
- Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Hu Dong
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Zhidong Teng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Sumin Wei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Yun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Shuanghui Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Jianli Tang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
- College of Animal Science, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
14
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [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: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
15
|
Newman NK, Zhang Y, Padiadpu J, Miranda CL, Magana AA, Wong CP, Hioki KA, Pederson JW, Li Z, Gurung M, Bruce AM, Brown K, Bobe G, Sharpton TJ, Shulzhenko N, Maier CS, Stevens JF, Gombart AF, Morgun A. Reducing gut microbiome-driven adipose tissue inflammation alleviates metabolic syndrome. MICROBIOME 2023; 11:208. [PMID: 37735685 PMCID: PMC10512512 DOI: 10.1186/s40168-023-01637-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/01/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND The gut microbiota contributes to macrophage-mediated inflammation in adipose tissue with consumption of an obesogenic diet, thus driving the development of metabolic syndrome. There is a need to identify and develop interventions that abrogate this condition. The hops-derived prenylated flavonoid xanthohumol (XN) and its semi-synthetic derivative tetrahydroxanthohumol (TXN) attenuate high-fat diet-induced obesity, hepatosteatosis, and metabolic syndrome in C57Bl/6J mice. This coincides with a decrease in pro-inflammatory gene expression in the gut and adipose tissue, together with alterations in the gut microbiota and bile acid composition. RESULTS In this study, we integrated and interrogated multi-omics data from different organs with fecal 16S rRNA sequences and systemic metabolic phenotypic data using a Transkingdom Network Analysis. By incorporating cell type information from single-cell RNA-seq data, we discovered TXN attenuates macrophage inflammatory processes in adipose tissue. TXN treatment also reduced levels of inflammation-inducing microbes, such as Oscillibacter valericigenes, that lead to adverse metabolic phenotypes. Furthermore, in vitro validation in macrophage cell lines and in vivo mouse supplementation showed addition of O. valericigenes supernatant induced the expression of metabolic macrophage signature genes that are downregulated by TXN in vivo. CONCLUSIONS Our findings establish an important mechanism by which TXN mitigates adverse phenotypic outcomes of diet-induced obesity and metabolic syndrome. TXN primarily reduces the abundance of pro-inflammatory gut microbes that can otherwise promote macrophage-associated inflammation in white adipose tissue. Video Abstract.
Collapse
Affiliation(s)
- N K Newman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Y Zhang
- School of Biological and Population Health Sciences, Nutrition Program, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- Present address: Oregon Health & Science University, Portland, OR, USA
| | - J Padiadpu
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - C L Miranda
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - A A Magana
- Department of Chemistry, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - C P Wong
- School of Biological and Population Health Sciences, Nutrition Program, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - K A Hioki
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
- Present address: UMASS, Amherst, MA, USA
| | - J W Pederson
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Z Li
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - M Gurung
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
- Present address: Children Nutrition Center, USDA, Little Rock, AR, USA
| | - A M Bruce
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - K Brown
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
- Chemical, Biological & Environmental Engineering, Oregon State University, Corvallis, OR, USA
| | - G Bobe
- Department of Animal Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - T J Sharpton
- Department of Microbiology, Department of Statistics, Oregon State University, Corvallis, OR, USA
| | - N Shulzhenko
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA.
| | - C S Maier
- Department of Chemistry, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - J F Stevens
- Department of Pharmaceutical Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - A F Gombart
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Corvallis, OR, USA.
| | - A Morgun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
16
|
Pachathundikandi SK, Tegtmeyer N, Backert S. Masking of typical TLR4 and TLR5 ligands modulates inflammation and resolution by Helicobacter pylori. Trends Microbiol 2023; 31:903-915. [PMID: 37012092 DOI: 10.1016/j.tim.2023.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
Helicobacter pylori is a paradigm of chronic bacterial infection and is associated with peptic ulceration and malignancies. H. pylori uses specific masking mechanisms to avoid canonical ligands from activating Toll-like receptors (TLRs), such as lipopolysaccharide (LPS) modification and specific flagellin sequences that are not detected by TLR4 and TLR5, respectively. Thus, it was believed for a long time that H. pylori evades TLR recognition as a crucial strategy for immune escape and bacterial persistence. However, recent data indicate that multiple TLRs are activated by H. pylori and play a role in the pathology. Remarkably, H. pylori LPS, modified through changes in acylation and phosphorylation, is mainly sensed by other TLRs (TLR2 and TLR10) and induces both pro- and anti-inflammatory responses. In addition, two structural components of the cag pathogenicity island-encoded type IV secretion system (T4SS), CagL and CagY, were shown to contain TLR5-activating domains. These domains stimulate TLR5 and enhance immunity, while LPS-driven TLR10 signaling predominantly activates anti-inflammatory reactions. Here, we discuss the specific roles of these TLRs and masking mechanisms during infection. Masking of typical TLR ligands combined with evolutionary shifting to other TLRs is unique for H. pylori and has not yet been described for any other species in the bacterial kingdom. Finally, we highlight the unmasked T4SS-driven activation of TLR9 by H. pylori, which mainly triggers anti-inflammatory responses.
Collapse
Affiliation(s)
- Suneesh Kumar Pachathundikandi
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Dept. of Biology, Chair of Microbiology, Staudtstr. 5, 91058 Erlangen, Germany; Babasaheb Bhimrao Ambedkar University, Dept. of Environmental Microbiology, School of Earth and Environmental Sciences, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Nicole Tegtmeyer
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Dept. of Biology, Chair of Microbiology, Staudtstr. 5, 91058 Erlangen, Germany
| | - Steffen Backert
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Dept. of Biology, Chair of Microbiology, Staudtstr. 5, 91058 Erlangen, Germany.
| |
Collapse
|
17
|
Feng S, Zhang C, Chen S, He R, Chao G, Zhang S. TLR5 Signaling in the Regulation of Intestinal Mucosal Immunity. J Inflamm Res 2023; 16:2491-2501. [PMID: 37337514 PMCID: PMC10276996 DOI: 10.2147/jir.s407521] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023] Open
Abstract
Toll-like receptor 5 (TLR5) is a pattern recognition receptor that specifically recognizes flagellin and consequently plays a crucial role in the control of intestinal homeostasis by activating innate and adaptive immune responses. TLR5 overexpression, on the other hand, might disrupt the intestinal mucosal barrier, which serves as the first line of defense against harmful microbes. The intestine symbiotic bacteria, mucous layer, intestinal epithelial cells (IECs), adherens junctions (such as tight junctions and peripheral membrane proteins), the intestinal mucosal immune system, and cytokines make up the intestinal mucosal barrier. Impaired barrier function has been linked to intestinal illnesses such as inflammatory bowel disease (IBD). IBD is a persistent non-specific inflammatory illness of the digestive system with an unknown cause. It is now thought to be linked to infection, environment, genes, immune system, and the gut microbiota. The significance of immunological dysfunction in IBD has received more attention in recent years. The purpose of this paper is to explore TLR5's position in the intestinal mucosal barrier and its relevance to IBD.
Collapse
Affiliation(s)
- Shuyan Feng
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Chi Zhang
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Shanshan Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, People’s Republic of China
| | - Ruonan He
- Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Guanqun Chao
- Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, 310018, People’s Republic of China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, People’s Republic of China
| |
Collapse
|
18
|
Li Y, Yao H, Liu S, Song D, Wu C, Zhang S, Gao Q, Zhang L. The role of flagellin F in Vibrio Parahaemolyticus-induced intestinal immunity and functional domain identification. Int J Biol Macromol 2023; 244:125404. [PMID: 37327919 DOI: 10.1016/j.ijbiomac.2023.125404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
The marine pathogen Vibrio parahaemolyticus has caused huge economic losses to aquaculture. Flagellin is a key bacterial virulence factor that induces an inflammatory response via activation of Toll-like receptor 5 (TLR5) signaling. Herein, to explore the inflammatory activity of V. parahaemolyticus flagellins (flaA, flaB, flaC, flaD, flaE, and flaF), we investigated their ability to induce apoptosis in a fish cell line. All six flagellins induced severe apoptosis. Moreover, treatment with V. parahaemolyticus flagellins increased TLR5 and myeloid differentiation factor 88 (MyD88) expression and the production of TNF-α and IL-8 significantly. This indicated that flagellins might induce a TLR5-meditated immune response via an MyD88-dependent pathway. FlaF exhibited the strongest immunostimulatory effect; therefore, the interaction between TLR5 and flaF was screened using the yeast two-hybrid system. A significant interaction between the two proteins was observed, indicating that flaF binds directly to TLR5. Finally, the amino acids that participate in the TLR5-flaF interaction were identified using molecular simulation, which indicated three binding sites. These results deepen our understanding of the immunogenic properties of flagellins from V. parahaemolyticus, which could be used for vaccine development in the future.
Collapse
Affiliation(s)
- Yang Li
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Hongqing Yao
- Songjiang Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 201699, PR China
| | - Sushuang Liu
- Department of Life Sciences and Health, School of Science and Engineering, Huzhou College, PR China
| | - Dawei Song
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Choufei Wu
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Shaoyong Zhang
- College of Life Science, Huzhou University, Huzhou 313000, PR China
| | - Quanxin Gao
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| | - Liqin Zhang
- College of Life Science, Huzhou University, Huzhou 313000, PR China.
| |
Collapse
|
19
|
Recombinant Domain of Flagellin Promotes In Vitro a Chemotactic Inflammatory Profile in Human Immune Cells Independently of a Dendritic Cell Phenotype. Molecules 2023; 28:molecules28052394. [PMID: 36903639 PMCID: PMC10005431 DOI: 10.3390/molecules28052394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Flagellin is the major component of the flagellum in gram-positive and -negative bacteria and is also the ligand for the Toll-like receptor 5 (TLR5). The activation of TLR5 promotes the expression of proinflammatory cytokines and chemokines and the subsequent activation of T cells. This study evaluated a recombinant domain from the amino-terminus D1 domain (rND1) of flagellin from Vibrio anguillarum, a fish pathogen, as an immunomodulator in human peripheral blood mononuclear cells (PBMCs) and monocyte-derived dendritic cells (MoDCs). We demonstrated that rND1 induced an upregulation of proinflammatory cytokines in PBMCs, characterized at the transcriptional level by an expression peak of 220-fold for IL-1β, 20-fold for IL-8, and 65-fold for TNF-α. In addition, at the protein level, 29 cytokines and chemokines were evaluated in the supernatant and were correlated with a chemotactic signature. MoDCs treated with rND1 showed low levels of co-stimulatory and HLA-DR molecules and kept an immature phenotype with a decreased phagocytosis of dextran. We probed that rND1 from a non-human pathogen promotes modulation in human cells, and it may be considered for further studies in adjuvant therapies based on pathogen-associated patterns (PAMPs).
Collapse
|
20
|
Duplicated Flagellins in Pseudomonas Divergently Contribute to Motility and Plant Immune Elicitation. Microbiol Spectr 2023; 11:e0362122. [PMID: 36629446 PMCID: PMC9927476 DOI: 10.1128/spectrum.03621-22] [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] [Indexed: 01/12/2023] Open
Abstract
Flagellins are the main constituents of the flagellar filaments that provide bacterial motility, chemotactic ability, and host immune elicitation ability. Although the functions of flagellins have been extensively studied in bacteria with a single flagellin-encoding gene, the function of multiple flagellin-encoding genes in a single bacterial species is largely unknown. Here, the model plant-growth-promoting bacterium Pseudomonas kilonensis F113 was used to decipher the divergent functions of duplicated flagellins. We demonstrate that the two flagellins (FliC-1 and FliC-2) in 12 Pseudomonas strains, including F113, are evolutionarily distinct. Only the fliC-1 gene but not the fliC-2 gene in strain F113 is responsible for flagellar biogenesis, motility, and plant immune elicitation. The transcriptional expression of fliC-2 was significantly lower than that of fliC-1 in medium and in planta, most likely due to variations in promoter activity. In silico prediction revealed that all fliC-2 genes in the 12 Pseudomonas strains have a poorly conserved promoter motif. Compared to the Flg22-2 epitope (relative to FliC-2), Flg22-1 (relative to FliC-1) induced stronger FLAGELLIN SENSING 2 (FLS2)-mediated microbe-associated molecular pattern-triggered immunity and significantly inhibited plant root growth. A change in the 19th amino acid in Flg22-2 reduced its binding affinity to the FLS2/brassinosteroid insensitive 1-associated kinase 1 complex. Also, Flg22-2 epitopes in the other 11 Pseudomonas strains were presumed to have low binding affinity due to the same change in the 19th amino acid. These findings suggest that Pseudomonas has evolved duplicate flagellins, with only FliC-1 contributing to motility and plant immune elicitation. IMPORTANCE Flagellins have emerged as important microbial patterns. This work focuses on flagellin duplication in some plant-associated Pseudomonas. Our findings on the divergence of duplicated flagellins provide a conceptual framework for better understanding the functional determinant flagellin and its peptide in multiple-flagellin plant-growth-promoting rhizobacteria.
Collapse
|
21
|
Kumar L, Brenner N, Sledzieski S, Olaosebikan M, Roger LM, Lynn-Goin M, Klein-Seetharaman R, Berger B, Putnam H, Yang J, Lewinski NA, Singh R, Daniels NM, Cowen L, Klein-Seetharaman J. Transfer of knowledge from model organisms to evolutionarily distant non-model organisms: The coral Pocillopora damicornis membrane signaling receptome. PLoS One 2023; 18:e0270965. [PMID: 36735673 PMCID: PMC9897584 DOI: 10.1371/journal.pone.0270965] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
With the ease of gene sequencing and the technology available to study and manipulate non-model organisms, the extension of the methodological toolbox required to translate our understanding of model organisms to non-model organisms has become an urgent problem. For example, mining of large coral and their symbiont sequence data is a challenge, but also provides an opportunity for understanding functionality and evolution of these and other non-model organisms. Much more information than for any other eukaryotic species is available for humans, especially related to signal transduction and diseases. However, the coral cnidarian host and human have diverged over 700 million years ago and homologies between proteins in the two species are therefore often in the gray zone, or at least often undetectable with traditional BLAST searches. We introduce a two-stage approach to identifying putative coral homologues of human proteins. First, through remote homology detection using Hidden Markov Models, we identify candidate human homologues in the cnidarian genome. However, for many proteins, the human genome alone contains multiple family members with similar or even more divergence in sequence. In the second stage, therefore, we filter the remote homology results based on the functional and structural plausibility of each coral candidate, shortlisting the coral proteins likely to have conserved some of the functions of the human proteins. We demonstrate our approach with a pipeline for mapping membrane receptors in humans to membrane receptors in corals, with specific focus on the stony coral, P. damicornis. More than 1000 human membrane receptors mapped to 335 coral receptors, including 151 G protein coupled receptors (GPCRs). To validate specific sub-families, we chose opsin proteins, representative GPCRs that confer light sensitivity, and Toll-like receptors, representative non-GPCRs, which function in the immune response, and their ability to communicate with microorganisms. Through detailed structure-function analysis of their ligand-binding pockets and downstream signaling cascades, we selected those candidate remote homologues likely to carry out related functions in the corals. This pipeline may prove generally useful for other non-model organisms, such as to support the growing field of synthetic biology.
Collapse
Affiliation(s)
- Lokender Kumar
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | - Nathanael Brenner
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | - Samuel Sledzieski
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Monsurat Olaosebikan
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Liza M. Roger
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Matthew Lynn-Goin
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | | | - Bonnie Berger
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Hollie Putnam
- Department of Biological Sciences, University of Rhode Island, South Kingstown, RI, United States of America
| | - Jinkyu Yang
- Department of Department of Aeronautics & Astronautics, University of Washington, Seattle, WA, United States of America
| | - Nastassja A. Lewinski
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rohit Singh
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Noah M. Daniels
- Department of Computer Science and Statistics, University of Rhode Island, South Kingstown, RI, United States of America
| | - Lenore Cowen
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Judith Klein-Seetharaman
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
- * E-mail:
| |
Collapse
|
22
|
In silico peptide-based therapeutics against human colorectal cancer by the activation of TLR5 signaling pathways. J Mol Model 2023; 29:35. [PMID: 36626012 DOI: 10.1007/s00894-022-05422-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in both men and women. Toll-like receptor 5 (TLR5), an autoimmune signaling receptor that plays a role in cancer, can be exploited for the suppression of human colon cancer. Salmonella flagellin protein, a novel agonist of TLR5 activating downstream signaling, could be a basis for designing anticancer peptides. METHODS The three-dimensional crystal structure of TLR5 (PDB ID: 3J0A, Resolution = 26.0 Å) was optimized using the AMBER force field in the YASARA suit. In silico enzymatic digestion tool, PeptideCutter, was used to identify peptides from Salmonella flagellin, an agonist against human TLR5. The 3D structure of the peptides was generated using PEP-FOLD3. These peptides were screened against human TLR5 using shape complementarity principles based on the binding affinity and interactions with the active residue of TLR5 monomer, and the selected peptides were further validated by molecular dynamic (MD) simulation. RESULTS In this study, we generated 42 peptides from Salmonella flagellin protein by in silico protein digestion. Then, based on a new hidden Markov model sub-optimal conformation sampling approach as well as the size of the fragments, we select 38 effective peptides from these 42 cleavages. These peptides were screened against the monomeric Xray structure of human TLR5 using shape complementarity principles. Based on the binding affinity and interactions with the active residue of TLR5 monomer (residues 294 and 366 of TLR5), nine top-scored peptides were selected for the initial molecular dynamic (MD) simulation. Among these peptides, Clv10, Clv17, and Clv28 showed high stability and less flexibility during MD simulation. A 1 μs MD simulation was performed on TLR5-Clv10, TLR-Clv17, and TLR5-Clv28 complexes to further analyze the stability, conformational changes, and binding mode (Clv10, Clv17, and Clv28). During this MD study, the peptides showed high salt bridges and ionic interactions with residue ASP294 and residue ASP366 throughout the simulation and remained in the concave of the human TLR5 monomer. The RMSD and Rg values showed that the peptide-protein complexes become stable after 200 ns of contraction and extraction. CONCLUSION These findings can facilitate the rational design of selected peptides as an agonist of TLR5, which have antitumor activity, suppress colorectal cancer tumors, and can be used as promising candidates and novel agonists of TLR5.
Collapse
|
23
|
Scheithauer L, Karagöz MS, Mayer BE, Steinert M. Protein sociology of ProA, Mip and other secreted virulence factors at the Legionella pneumophila surface. Front Cell Infect Microbiol 2023; 13:1140688. [PMID: 36936764 PMCID: PMC10017501 DOI: 10.3389/fcimb.2023.1140688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The pathogenicity of L. pneumophila, the causative agent of Legionnaires' disease, depends on an arsenal of interacting proteins. Here we describe how surface-associated and secreted virulence factors of this pathogen interact with each other or target extra- and intracellular host proteins resulting in host cell manipulation and tissue colonization. Since progress of computational methods like AlphaFold, molecular dynamics simulation, and docking allows to predict, analyze and evaluate experimental proteomic and interactomic data, we describe how the combination of these approaches generated new insights into the multifaceted "protein sociology" of the zinc metalloprotease ProA and the peptidyl-prolyl cis/trans isomerase Mip (macrophage infectivity potentiator). Both virulence factors of L. pneumophila interact with numerous proteins including bacterial flagellin (FlaA) and host collagen, and play important roles in virulence regulation, host tissue degradation and immune evasion. The recent progress in protein-ligand analyses of virulence factors suggests that machine learning will also have a beneficial impact in early stages of drug discovery.
Collapse
Affiliation(s)
- Lina Scheithauer
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Mustafa Safa Karagöz
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Benjamin E. Mayer
- Computational Biology & Simulation, Technische Universität Darmstadt, Darmstadt, Germany
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
- *Correspondence: Michael Steinert,
| |
Collapse
|
24
|
Yoon HJ, Lee S, Kim TY, Yu SE, Kim HS, Chung YS, Chung S, Park S, Shin YC, Wang EK, Noh J, Kim HJ, Ku CR, Koh H, Kim CS, Park JS, Shin YM, Sung HJ. Sprayable nanomicelle hydrogels and inflammatory bowel disease patient cell chips for development of intestinal lesion-specific therapy. Bioact Mater 2022; 18:433-445. [PMID: 35415304 PMCID: PMC8971598 DOI: 10.1016/j.bioactmat.2022.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/07/2022] [Accepted: 03/20/2022] [Indexed: 12/16/2022] Open
Abstract
All-in-one treatments represent a paradigm shift in future medicine. For example, inflammatory bowel disease (IBD) is mainly diagnosed by endoscopy, which could be applied for not only on-site monitoring but also the intestinal lesion-targeted spray of injectable hydrogels. Furthermore, molecular conjugation to the hydrogels would program both lesion-specific adhesion and drug-free therapy. This study validated this concept of all-in-one treatment by first utilizing a well-known injectable hydrogel that underwent efficient solution-to-gel transition and nanomicelle formation as a translatable component. These properties enabled spraying of the hydrogel onto the intestinal walls during endoscopy. Next, peptide conjugation to the hydrogel guided endoscopic monitoring of IBD progress upon adhesive gelation with subsequent moisturization of inflammatory lesions, specifically by nanomicelles. The peptide was designed to mimic the major component that mediates intestinal interaction with Bacillus subtilis flagellin during IBD initiation. Hence, the peptide-guided efficient adhesion of the hydrogel nanomicelles onto Toll-like receptor 5 (TLR5) as the main target of flagellin binding and Notch-1. The peptide binding potently suppressed inflammatory signaling without drug loading, where TLR5 and Notch-1 operated collaboratively through downstream actions of tumor necrosis factor-alpha. The results were produced using a human colorectal cell line, clinical IBD patient cells, gut-on-a-chip, a mouse IBD model, and pig experiments to validate the translational utility. Injectable nanomicelle hydrogel for all-in-one treatment of intestinal inflammation. Spraying of the hydrogel onto the intestinal walls during endoscopy. Peptide-guided detection and moisturization of inflammatory lesions.
Collapse
Affiliation(s)
- Hyo-Jin Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Songhyun Lee
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Tae Young Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eun Yu
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Young Shin Chung
- Department of Obstetrics and Gynecology, Institution of Women's Life Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seyong Chung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Suji Park
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eun Kyung Wang
- Department of Internal Medicine, Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jihye Noh
- Department of Pediatrics, Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyun Jung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Cheol Ryong Ku
- Department of Internal Medicine, Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hong Koh
- Department of Pediatrics, Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chang-Soo Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Numais Co., Ltd., Korea Seoul 04799, Republic of Korea
| | - Joon-Sang Park
- Department of Computer Engineering, Hongik University, Seoul, 04066, Republic of Korea
- Corresponding author.
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Corresponding author.
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Corresponding author.
| |
Collapse
|
25
|
Legionella pneumophila PPIase Mip Interacts with the Bacterial Proteins SspB, Lpc2061, and FlaA and Promotes Flagellation. Infect Immun 2022; 90:e0027622. [PMID: 36314784 PMCID: PMC9670971 DOI: 10.1128/iai.00276-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The peptidyl-prolyl-
cis/trans
-isomerase (PPIase) macrophage infectivity potentiator (Mip) contributes to the pathogenicity and fitness of
L. pneumophila
, the causative agent of Legionnaires’ disease. Here, we identified the stringent starvation protein SspB, hypothetical protein Lpc2061, and flagellin FlaA as bacterial interaction partners of Mip.
Collapse
|
26
|
Murtaza A, Afzal H, Doan TD, Ke GM, Cheng LT. Flagellin Improves the Immune Response of an Infectious Bursal Disease Virus (IBDV) Subunit Vaccine. Vaccines (Basel) 2022; 10:1780. [PMID: 36366289 PMCID: PMC9695526 DOI: 10.3390/vaccines10111780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 09/17/2024] Open
Abstract
Flagellin activates the immune system through Toll-like receptor 5 (TLR5) and can work as an adjuvant for subunit vaccines. In this study, we tested the adjuvancy of two different N-terminal fragments of flagellin, (1) FliC99, residues 1-99, and (2) FliC176, residues 1-176, to incorporate larger areas of the hotspot region for potentially higher levels of TLR5 activation and immune response. A truncated version of the VP2 protein (name tVP2, residues 199-356) of the Infectious bursal disease virus (IBDV) was genetically linked to the flagellin constructs, and the immune response was evaluated in chickens. Results showed that both chimeric antigen-adjuvant constructs increased humoral (total IgG titers), cellular and cytokine immune response (IL-4, IFN-γ). The resulting antibody also successfully neutralized IBDV. We conclude that the N-terminus of flagellin can act as an immune activator to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Asad Murtaza
- International Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Centre for Immunology & Infection, Hong Kong Science and Technology Park, Hong Kong, China
| | - Haroon Afzal
- International Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Thu-Dung Doan
- International Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- General Research Service Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Guan-Ming Ke
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| |
Collapse
|
27
|
Genome-Wide Identification and Characterization of Toll-like Receptors (TLRs) in Diaphorina citri and Their Expression Patterns Induced by the Endophyte Beauveria bassiana. J Fungi (Basel) 2022; 8:jof8080888. [PMID: 36012876 PMCID: PMC9409752 DOI: 10.3390/jof8080888] [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: 07/29/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Toll-like receptors (TLRs) are pathogen recognition receptors (PRRs), which play key roles in helping the host immune system fight pathogen invasions. Systematic information on TLRs at the genome-wide level and expression profiling in response to endophytic colonization is very important to understand their functions but is currently lacking in this field. Here, a total of two TLR genes were identified and characterized in Diaphorina citri. The TLR genes of D. citri were clustered into five families according to the phylogenetic analysis of different species' TLRs. The domain organization analyses suggested that the TLRs were constituted of three important parts: a leucine-rich repeat (LRR) domain, a transmembrane region (TR) and a Toll/interleukin-1 receptor (TIR) domain. The mRNA expression levels of the two TLR genes (DcTOLL and DcTLR7) were highly regulated in both nymphs and adults of D. citri. These results elucidated the potentiated TLR gene expression in response to endophytically colonized plants. Furthermore, the 3D structures of the TIR domain were highly conserved during evolution. Collectively, these findings elucidate the crucial roles of TLRs in the immune response of D. citri to entomopathogens systematically established as endophytes, and provide fundamental knowledge for further understanding of the innate immunity of D. citri.
Collapse
|
28
|
Liao Z, Yang C, Jiang R, Zhu W, Zhang Y, Su J. Cyprinid-specific duplicated membrane TLR5 senses dsRNA as functional homodimeric receptors. EMBO Rep 2022; 23:e54281. [PMID: 35678424 DOI: 10.15252/embr.202154281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 05/02/2022] [Accepted: 05/13/2022] [Indexed: 12/30/2022] Open
Abstract
Membrane-embedded Toll-like receptor 5 (TLR5) functions as a homodimer to detect bacterial flagellin. Cyprinid grass carp (Ctenopharyngodon idella) encodes two TLR5 genes, CiTLR5a and CiTLR5b. Here, we show that cyprinid TLR5a and TLR5b homodimers unexpectedly bind the dsRNA analog poly(I:C) and regulate interferon (IFN) response in early endosomes and lysosomes. Although TLR5 homodimers also bind flagellin, an immune response to flagellin is only triggered by TLR5a/b heterodimer. Moreover, we demonstrate that two TLR5 paralogs have opposite effects on antiviral response: CiTLR5a slightly promotes and powerfully maintains, whereas CiTLR5b remarkably inhibits virus replication. We show that the ectodomain of CiTLR5 is required for dsRNA-induced IFN signaling, and we map the key poly(I:C) binding sites to G240 for CiTLR5a and to N547 for CiTLR5b. Furthermore, we reveal that differential N-glycosylation of CiTLR5a/b affects dsRNA-IFN signaling but has no role in flagellin-mediated NF-κB induction, with paralog-specific roles for CiTLR5a-T101 and corresponding CiTLR5b-I99. Moreover, we provide evidence that the ability to sense dsRNA represents a neofunctionalization specific for membrane-bound TLR5 in cyprinid, bridging viral and bacterial immune responses.
Collapse
Affiliation(s)
- Zhiwei Liao
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Jiang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Wentao Zhu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Yongan Zhang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
29
|
Shao H, Min F, Huang M, Wang Z, Bai T, Lin M, Li X, Chen H. Novel perspective on the regulation of food allergy by probiotic: The potential of its structural components. Crit Rev Food Sci Nutr 2022; 64:172-186. [PMID: 35912422 DOI: 10.1080/10408398.2022.2105304] [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] [Indexed: 11/03/2022]
Abstract
Food allergy (FA) is a global public health issue with growing prevalence. Increasing evidence supports the strong correlation between intestinal microbiota dysbiosis and food allergies. Probiotic intervention as a microbiota-based therapy could alleviate FA effectively. In addition to improving the intestinal microbiota disturbance and affecting microbial metabolites to regulate immune system, immune responses induced by the recognition of pattern recognition receptors to probiotic components may also be one of the mechanisms of probiotics protecting against FA. In this review, it is highlighted in detail about the regulatory effects on the immune system and anti-allergic potential of probiotic components including the flagellin, pili, peptidoglycan, lipoteichoic acid, exopolysaccharides, surface (S)-layer proteins and DNA. Probiotic components could enhance the function of intestinal epithelial barrier as well as regulate the balance of cytokines and T helper (Th) 1/Th2/regulatory T cell (Treg) responses. These evidences suggest that probiotic components could be used as nutritional or therapeutic agents for maintaining immune homeostasis to prevent FA, which will contribute to providing new insights into the resolution of FA and better guidance for the development of probiotic products.
Collapse
Affiliation(s)
- Huming Shao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Fangfang Min
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Meijia Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Zhongliang Wang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Tianliang Bai
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Min Lin
- Department of Dermatology, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- School of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, Jiangxi, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
30
|
Chuekwon K, Chu CY, Cheng LT. N-terminus of flagellin enhances vaccine efficacy against Actinobacillus pleuropneumoniae. BMC Vet Res 2022; 18:279. [PMID: 35842618 PMCID: PMC9288005 DOI: 10.1186/s12917-022-03380-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Flagellin elicits potent immune response and may serve as a vaccine adjuvant. We previously reported that the N-terminus of flagellin (residues 1-99, nFliC) is sufficient for vaccine efficacy enhancement against Pasteurella multocida challenge in chickens. In this study, we futher tested the adjuvancy of nFliC in a subunit vaccine against the pig pathogen Actinobacillus pleuropneumoniae in a mice model. For vaccine formulation, the antigen ApxIIPF (the pore-forming region of the exotoxin ApxII) was combined with nFliC, either through genetic fusion or simple admixture. RESULTS Immune analysis showed that nFliC, introduced through genetic fusion or admixture, enhanced both humoral (antibody levels) and cellular (T cell response and cytokine production) immunity. In a challenge test, nFliC increased vaccine protective efficacy to 60-80%, vs. 20% for the antigen-only group. Further analysis showed that, even without a supplemental adjuvant such as mineral salt or oil emulsion, genetically linked nFliC still provided significant immune enhancement. CONCLUSIONS We conclude that nFliC is a versatile and potent adjuvant for vaccine formulation.
Collapse
Affiliation(s)
- Kamonpun Chuekwon
- Department of Tropical Agriculture and International Cooperation, International College, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan
| | - Chun-Yen Chu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung, 91201, Taiwan.
| |
Collapse
|
31
|
Sharp C, Foster KR. Host control and the evolution of cooperation in host microbiomes. Nat Commun 2022; 13:3567. [PMID: 35732630 PMCID: PMC9218092 DOI: 10.1038/s41467-022-30971-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Humans, and many other species, are host to diverse symbionts. It is often suggested that the mutual benefits of host-microbe relationships can alone explain cooperative evolution. Here, we evaluate this hypothesis with evolutionary modelling. Our model predicts that mutual benefits are insufficient to drive cooperation in systems like the human microbiome, because of competition between symbionts. However, cooperation can emerge if hosts can exert control over symbionts, so long as there are constraints that limit symbiont counter evolution. We test our model with genomic data of two bacterial traits monitored by animal immune systems. In both cases, bacteria have evolved as predicted under host control, tending to lose flagella and maintain butyrate production when host-associated. Moreover, an analysis of bacteria that retain flagella supports the evolution of host control, via toll-like receptor 5, which limits symbiont counter evolution. Our work puts host control mechanisms, including the immune system, at the centre of microbiome evolution.
Collapse
Affiliation(s)
- Connor Sharp
- Department of Zoology, University of Oxford, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Kevin R Foster
- Department of Zoology, University of Oxford, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
32
|
Dai F, Guo M, Shao Y, Li C. Vibrio splendidus flagellin C binds tropomodulin to induce p38 MAPK-mediated p53-dependent coelomocyte apoptosis in Echinodermata. J Biol Chem 2022; 298:102091. [PMID: 35654141 PMCID: PMC9249833 DOI: 10.1016/j.jbc.2022.102091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 12/27/2022] Open
Abstract
As a typical pathogen-associated molecular pattern, bacterial flagellin can bind Toll-like receptor 5 and the intracellular NAIP5 receptor component of the NLRC4 inflammasome to induce immune responses in mammals. However, these flagellin receptors are generally poorly understood in lower animal species. In this study, we found that the isolated flagellum of Vibrio splendidus AJ01 destroyed the integrity of the tissue structure of coelomocytes and promoted apoptosis in the sea cucumber Apostichopus japonicus. To further investigate the molecular mechanism, the novel intracellular LRR domain-containing protein tropomodulin (AjTmod) was identified as a protein that interacts with flagellin C (FliC) with a dissociation constant (Kd) of 0.0086 ± 0.33 μM by microscale thermophoresis assay. We show that knockdown of AjTmod also depressed FliC-induced apoptosis of coelomocytes. Further functional analysis with different inhibitor treatments revealed that the interaction between AjTmod and FliC could specifically activate p38 MAPK, but not JNK or ERK MAP kinases. We demonstrate that the transcription factor p38 is then translocated into the nucleus, where it mediates the expression of p53 to induce coelomocyte apoptosis. Our findings provide the first evidence that intracellular AjTmod serves as a novel receptor of FliC and mediates p53-dependent coelomocyte apoptosis by activating the p38 MAPK signaling pathway in Echinodermata.
Collapse
Affiliation(s)
- Fa Dai
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Yina Shao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, China; State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, PR China.
| |
Collapse
|
33
|
Scheithauer L, Thiem S, Ünal CM, Dellmann A, Steinert M. Zinc Metalloprotease ProA from Legionella pneumophila Inhibits the Pro-Inflammatory Host Response by Degradation of Bacterial Flagellin. Biomolecules 2022; 12:624. [PMID: 35625552 PMCID: PMC9138289 DOI: 10.3390/biom12050624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
The environmental bacterium Legionella pneumophila is an intracellular pathogen of various protozoan hosts and able to cause Legionnaires' disease, a severe pneumonia in humans. By encoding a wide selection of virulence factors, the infectious agent possesses several strategies to manipulate its host cells and evade immune detection. In the present study, we demonstrate that the L. pneumophila zinc metalloprotease ProA functions as a modulator of flagellin-mediated TLR5 stimulation and subsequent activation of the pro-inflammatory NF-κB pathway. We found ProA to be capable of directly degrading immunogenic FlaA monomers but not the polymeric form of bacterial flagella. These results indicate a role of the protease in antagonizing immune stimulation, which was further substantiated in HEK-BlueTM hTLR5 Detection assays. Addition of purified proteins, bacterial suspensions of L. pneumophila mutant strains as well as supernatants of human lung tissue explant infection to this reporter cell line demonstrated that ProA specifically decreases the TLR5 response via FlaA degradation. Conclusively, the zinc metalloprotease ProA serves as a powerful regulator of exogenous flagellin and presumably creates an important advantage for L. pneumophila proliferation in mammalian hosts by promoting immune evasion.
Collapse
Affiliation(s)
- Lina Scheithauer
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Stefanie Thiem
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Can M. Ünal
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Ansgar Dellmann
- Institut für Pathologie, Städtisches Klinikum Braunschweig, Celler Straße 38, 38114 Braunschweig, Germany;
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
- Helmholtz Center for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
34
|
Shen M, Jiang Z, Zhang K, Li C, Liu F, Hu Y, Zheng S, Zheng R. Transcriptome analysis of grass carp (Ctenopharyngodon idella) and Holland's spinibarbel (Spinibarbus hollandi) infected with Ichthyophthirius multifiliis. FISH & SHELLFISH IMMUNOLOGY 2022; 121:305-315. [PMID: 35031476 DOI: 10.1016/j.fsi.2022.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Ichthyophthirius multifiliis is a protozoan ciliate that causes white spot disease (also known as ichthyophthiriasis) in freshwater fish. Holland's spinibarbel (Spinibarbus hollandi) was less susceptible to white spot disease than grass carp (Ctenopharyngodon Idella). In this study, grass carp and Holland's spinibarbel are infected by I. multifiliis and the amount of infection is 10,000 theronts per fish. All grass carp died within 12 days after infection, and the survival rate of Holland's spinibarbel was more than 80%. In order to study the difference in sensitivity of these two fish species to I. multifiliis, transcriptome analysis was conducted using gill, skin, liver, spleen and head kidney of Holland's spinibarbel and grass carp at 48 h post-infection with I. multifiliis. A total of 489,296,696 clean reads were obtained by sequencing. A total of 105 significantly up-regulated immune-related genes were obtained by Gene Ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis in grass carp. Cluster of differentiation 40 (CD40), cluster of differentiation 80 (CD 80), tumor necrosis factor-alpha (TNF-α), toll-like receptor 4 (TLR-4), interleukin 1 beta (IL-1β) and other inflammatory-related genes in grass carp were enriched in the cytokine-cytokine receptor interaction pathway and toll-like receptor pathway. In Holland's spinibarbel, a total of 46 significantly up-regulated immune-related genes were obtained by GO classification and KEGG pathway enrichment analysis. Immune-related genes, such as Immunoglobin heavy chain (IgH), cathepsin S (CTSS), complement C1q A chain (C1qA), complement component 3 (C3) and complement component (C9) were enriched in phagosome pathway, lysosome pathway and complement and coagulation concatenation pathway. C3 was significantly up-regulated in gill and head kidney. Fluorescence in situ hybridization (FISH) showed that the C3 gene was highly expressed in gill tissue of Holland's spinibarbel infected with I. multifiliis. A small amount of C3 gene was expressed in the gill arch of grass carp after infected with I. multifiliis. In conclusion, the severe inflammatory response in vivo after infecting grass carp with I. multifiliis might be the main cause of the death of grass carp. The extrahepatic expression of the gene of Holland's spinibarbel might play an important role in the immune defense against I. multifiliis.
Collapse
Affiliation(s)
- Minghao Shen
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Zeyuan Jiang
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Kai Zhang
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Chenyang Li
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Fangling Liu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Yibing Hu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Shanjian Zheng
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China.
| | - Rongquan Zheng
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, 321004, China; Key Lab of Wildlife Biotechnology and Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua, 321004, China; Xinzhi College, Zhejiang Normal University, Jinhua, 321004, China.
| |
Collapse
|
35
|
Côté-Cyr M, Zottig X, Gauthier L, Archambault D, Bourgault S. Self-Assembly of Flagellin into Immunostimulatory Ring-like Nanostructures as an Antigen Delivery System. ACS Biomater Sci Eng 2022; 8:694-707. [PMID: 35080372 DOI: 10.1021/acsbiomaterials.1c01332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Proteinaceous nanoparticles represent attractive antigen carriers for vaccination as their size and repetitive antigen displays that mimic most viral particles enable efficient immune processing. However, these nanocarriers are often unable to stimulate efficiently the innate immune system, requiring coadministration with adjuvants to promote long-lasting protective immunity. The protein flagellin, which constitutes the primary constituent of the bacterial flagellum, has been widely evaluated as an antigen carrier due to its intrinsic adjuvant properties involving activation of the innate immune receptor Toll-like receptor 5 (TLR5). Although flagellin is known for its ability to self-assemble into micron-scale length nanotubes, few studies have evaluated the potential usage of flagellin-based nanostructures as immunostimulatory antigen carriers. In this study, we reported for the first time a strategy to guide the self-assembly of a flagellin protein from Bacillus subtilis, Hag, into lower aspect ratio nanoparticles by hindering non-covalent interactions responsible for its elongation into nanotubes. We observed that addition of an antigenic sequence derived from the influenza A virus (3M2e) at the C-terminus of this flagellin, as opposed to positioning the epitope into mid-sequence, precluded filament elongation and resulted in low aspect ratio ring-like nanostructures upon salting-out-induced self-assembly. These nanostructures displayed the antigen at their surface and shared morphological and structural characteristics with flagellin nanotubes, with a diameter of approximately 12 nm, and an α-helix-rich secondary structure. Flagellin ring-like nanostructures were efficiently internalized by antigen-presenting cells, and avidly activated the TLR5 in vitro as well as the innate and adaptive immune responses. Intranasal immunization of mice with these nanostructures resulted in the potentiation of the antigen-specific antibody response and protection against a lethal infection with the influenza A virus, illustrating the potential of these intrinsically immunostimulatory nanostructures as antigen carriers.
Collapse
Affiliation(s)
- Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Laurie Gauthier
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| |
Collapse
|
36
|
Li H, Zhang H, Huang G, Bing Z, Xu D, Liu J, Luo H, An X. Loss of RPS27a expression regulates the cell cycle, apoptosis, and proliferation via the RPL11-MDM2-p53 pathway in lung adenocarcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:33. [PMID: 35073964 PMCID: PMC8785590 DOI: 10.1186/s13046-021-02230-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Background Depletion of certain ribosomal proteins induces p53 activation, which is mediated mainly by ribosomal protein L5 (RPL5) and/or ribosomal protein L11 (RPL11). Therefore, RPL5 and RPL11 may link RPs and p53 activation. Thus, this study aimed to explore whether RPs interact with RPL11 and regulate p53 activation in lung adenocarcinoma (LUAD) cells. Methods The endogenous RPL11-binding proteins in A549 cells were pulled down through immunoprecipitation and identified with a proteomics approach. Docking analysis and GST-fusion protein assays were used to analyze the interaction of ribosomal protein S27a (RPS27a) and RPL11. Co-immunoprecipitation and in vitro ubiquitination assays were used to detect the effects of knockdown of RPS27a on the interaction between RPS27a and RPL11, and on p53 accumulation. Cell cycle, apoptosis, cell invasion and migration, cell viability and colony-formation assays were performed in the presence of knockdown of RPS27a. The RPS27a mRNA expression in LUAD was analyzed on the basis of the TCGA dataset, and RPS27a expression was detected through immunohistochemistry in LUAD samples. Finally, RPS27a and p53 expression was analyzed through immunohistochemistry in A549 cell xenografts with knockdown of RPS27a. Results RPS27a was identified as a novel RPL11 binding protein. GST pull-down assays revealed that RPS27a directly bound RPL11. Knockdown of RPS27a weakened the interaction between RPS27a and RPL11, but enhanced the binding of RPL11 and murine double minute 2 (MDM2), thereby inhibiting the ubiquitination and degradation of p53 by MDM2. Knockdown of RPS27a stabilized p53 in an RPL11-dependent manner and induced cell viability inhibition, cell cycle arrest and apoptosis in a p53-dependent manner in A549 cells. The expression of RPS27a was upregulated in LUAD and correlated with LUAD progression and poorer prognosis. Overexpression of RPS27a correlated with upregulation of p53, MDM2 and RPL11 in LUAD clinical specimens. Knockdown of RPS27a increased p53 activation, thus, suppressing the formation of A549 cell xenografts in nude mice. Conclusions RPS27a interacts with RPL11, and RPS27a knockdown enhanced the binding of RPL11 and MDM2, thereby inhibiting MDM2-mediated p53 ubiquitination and degradation; in addition, RPS27a as important roles in LUAD progression and prognosis, and may be a therapeutic target for patients with LUAD. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02230-z.
Collapse
|
37
|
Côté-Cyr M, Gauthier L, Zottig X, Bourgault S, Archambault D. Recombinant Bacillus subtilis flagellin Hag is a potent immunostimulant with reduced proinflammatory properties compared to Salmonella enterica serovar Typhimurium FljB. Vaccine 2022; 40:11-17. [PMID: 34844822 DOI: 10.1016/j.vaccine.2021.11.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022]
Abstract
Flagellin constitutes a potential adjuvant for vaccines owing to its robust immunostimulatory properties. However, clinical trials have revealed that flagellin derived from Salmonella enterica serovar Typhimurium induces high levels of proinflammatory markers and substantial adverse effects. The flagellin from Bacillus subtilis, Hag, shares high sequence homology with Salmonella FljB within the D0 and D1 domains responsible for TLR5 engagement, while the D2 and D3 domains associated with an off-target immune response are absent. Accordingly, we compared the immunostimulatory and proinflammatory properties of Hag with FljB by harnessing an epitope from the matrix 2 protein (M2e) of the influenza virus. Both flagellins engaged TLR5, with FljB showing a 2.5-fold higher potency than Hag. Mice inoculation showed a robust FljB- or Hag-induced M2e-specific antibody response, with Hag demonstrating a decreased secretion of proinflammatory markers and reduced weight loss. This study revealed that flagellin Hag is a potent immunoadjuvant with reduced proinflammatory properties.
Collapse
Affiliation(s)
- Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Laurie Gauthier
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montréal, Canada; Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada.
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Canada; The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe, Canada.
| |
Collapse
|
38
|
Gao Q, Yi S, Li Y, Luo J, Xing Q, Yang X, Zhao M, Min M, Wang Q, Wang Y, Ma L, Peng S. The Role of Flagellin B in Vibrio anguillarum-Induced Intestinal Immunity and Functional Domain Identification. Front Immunol 2021; 12:774233. [PMID: 34912344 PMCID: PMC8667730 DOI: 10.3389/fimmu.2021.774233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Vibrio anguillarum, an opportunistic pathogen of aquatic animals, moves using a filament comprised of polymerised flagellin proteins. Flagellins are essential virulence factors for V. anguillarum infection. Herein, we investigated the effects of flagellins (flaA, flaB, flaC, flaD and flaE) on cell apoptosis, TLR5 expression, and production of IL-8 and TNF-α. FlaB exhibited the strongest immunostimulation effects. To explore the functions of flaB in infection, we constructed a flaB deletion mutant using a two-step recombination method, and in vitro experiments showed a significant decrease in the expression of TLR5 and inflammatory cytokines compared with wild-type cells. However in the in vivo study, expression of inflammatory cytokines and intestinal mucosal structure showed no significant differences between groups. Additionally, flaB induced a significant increase in TLR5 expression based on microscopy analysis of fluorescently labelled TLR5, indicating interactions between the two proteins, which was confirmed by native PAGE and yeast two-hybrid assay. Molecular simulation of interactions between flaB and TLR5 was performed to identify the residues involved in binding, revealing two binding sites. Then, based on molecular dynamics simulations, we carried out thirteen site-directed mutations occurring at the amino acid sites of Q57, N83, N87, R91, D94, E122, D152, N312, R313, N320, L97, H316, I324 in binding regions of flaB protein by TLR5, respectively. Surface plasmon resonance (SPR) was employed to compare the affinities of flaB mutants for TLR5, and D152, D94, I324, N87, R313, N320 and H316 were found to mediate interactions between flaB and TLR5. Our comprehensive and systematic analysis of V. anguillarum flagellins establishes the groundwork for future design of flagellin-based vaccines.
Collapse
Affiliation(s)
- Quanxin Gao
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Shaokui Yi
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Yang Li
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Jinping Luo
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Qianqian Xing
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Xia Yang
- Zhejiang Provincial Key Laboratory of Aquatic Resources, Conservation and Development, Key Laboratory of Aquatic Animal Genetic Breeding and Nutrition, Chinese Academy of Fishery Sciences, College of Life Science, Huzhou University, Huzhou, China
| | - Ming Zhao
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Minghua Min
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Qian Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Yabing Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Lingbo Ma
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Shiming Peng
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| |
Collapse
|
39
|
Pandey M, Bhati A, Priya K, Sharma KK, Singhal B. Precision Postbiotics and Mental Health: the Management of Post-COVID-19 Complications. Probiotics Antimicrob Proteins 2021; 14:426-448. [PMID: 34806151 PMCID: PMC8606251 DOI: 10.1007/s12602-021-09875-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/14/2023]
Abstract
The health catastrophe originated by COVID-19 pandemic construed profound impact on a global scale. However, a plethora of research studies corroborated convincing evidence conferring severity of infection of SARS-CoV-2 with the aberrant gut microbiome that strongly speculated its importance for development of novel therapeutic modalities. The intense exploration of probiotics has been envisaged to promote the healthy growth of the host, and restore intestinal microecological balance through various metabolic and physiological processes. The demystifying effect of probiotics cannot be defied, but there exists a strong skepticism related to their safety and efficacy. Therefore, molecular signature of probiotics termed as "postbiotics" are of paramount importance and there is continuous surge of utilizing postbiotics for enhancing health benefits, but little is explicit about their antiviral effects. Therefore, it is worth considering their prospective role in post-COVID regime that pave the way for exploring the pastoral vistas of postbiotics. Based on previous research investigations, the present article advocates prospective role of postbiotics in alleviating the health burden of viral infections, especially SARS-CoV-2. The article also posits current challenges and proposes a futuristic model describing the concept of "precision postbiotics" for effective therapeutic and preventive interventions that can be used for management of this deadly disease.
Collapse
Affiliation(s)
- Muskan Pandey
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Archana Bhati
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Kumari Priya
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - K K Sharma
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Barkha Singhal
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India.
| |
Collapse
|
40
|
Sartorius R, Trovato M, Manco R, D'Apice L, De Berardinis P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines 2021; 6:127. [PMID: 34711839 PMCID: PMC8553822 DOI: 10.1038/s41541-021-00391-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are transmembrane proteins belonging to the family of pattern-recognition receptors. They function as sensors of invading pathogens through recognition of pathogen-associated molecular patterns. After their engagement by microbial ligands, TLRs trigger downstream signaling pathways that culminate into transcriptional upregulation of genes involved in immune defense. Here we provide an updated overview on members of the TLR family and we focus on their role in antiviral response. Understanding of innate sensing and signaling of viruses triggered by these receptors would provide useful knowledge to prompt the development of vaccines able to elicit effective and long-lasting immune responses. We describe the mechanisms developed by viral pathogens to escape from immune surveillance mediated by TLRs and finally discuss how TLR/virus interplay might be exploited to guide the design of innovative vaccine platforms.
Collapse
Affiliation(s)
- Rossella Sartorius
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Roberta Manco
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Luciana D'Apice
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | | |
Collapse
|
41
|
Nassar A, Ibrahim IM, Amin FG, Magdy M, Elgharib AM, Azzam EB, Nasser F, Yousry K, Shamkh IM, Mahdy SM, Elfiky AA. A Review of Human Coronaviruses' Receptors: The Host-Cell Targets for the Crown Bearing Viruses. Molecules 2021; 26:6455. [PMID: 34770863 PMCID: PMC8587140 DOI: 10.3390/molecules26216455] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/19/2022] Open
Abstract
A novel human coronavirus prompted considerable worry at the end of the year 2019. Now, it represents a significant global health and economic burden. The newly emerged coronavirus disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the primary reason for the COVID-19 global pandemic. According to recent global figures, COVID-19 has caused approximately 243.3 million illnesses and 4.9 million deaths. Several human cell receptors are involved in the virus identification of the host cells and entering them. Hence, understanding how the virus binds to host-cell receptors is crucial for developing antiviral treatments and vaccines. The current work aimed to determine the multiple host-cell receptors that bind with SARS-CoV-2 and other human coronaviruses for the purpose of cell entry. Extensive research is needed using neutralizing antibodies, natural chemicals, and therapeutic peptides to target those host-cell receptors in extremely susceptible individuals. More research is needed to map SARS-CoV-2 cell entry pathways in order to identify potential viral inhibitors.
Collapse
Affiliation(s)
- Aaya Nassar
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Ibrahim M. Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Fatma G. Amin
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
- Physics Department, Faculty of Science, Alexandria University, Alexandria 21519, Egypt
| | - Merna Magdy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Ahmed M. Elgharib
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| | - Eman B. Azzam
- Physics Department, Medical Biophysics Division, Faculty of Science, Helwan University, Cairo 11511, Egypt;
| | - Filopateer Nasser
- Biochemistry Department, Faculty of Science, Cairo University, Giza 12511, Egypt;
| | - Kirllos Yousry
- Faculty of Medicine, Cairo University, Cairo 11511, Egypt;
| | | | - Samah M. Mahdy
- National Museum of Egyptian Civilization, Ain Elsira-Elfustat, Cairo 11511, Egypt;
| | - Abdo A. Elfiky
- Biophysics Department, Faculty of Science, Cairo University, Giza 12511, Egypt; (I.M.I.); (F.G.A.); (M.M.); (A.M.E.)
| |
Collapse
|
42
|
Pogostin BH, McHugh KJ. Novel Vaccine Adjuvants as Key Tools for Improving Pandemic Preparedness. Bioengineering (Basel) 2021; 8:155. [PMID: 34821721 PMCID: PMC8615241 DOI: 10.3390/bioengineering8110155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Future infectious disease outbreaks are inevitable; therefore, it is critical that we maximize our readiness for these events by preparing effective public health policies and healthcare innovations. Although we do not know the nature of future pathogens, antigen-agnostic platforms have the potential to be broadly useful in the rapid response to an emerging infection-particularly in the case of vaccines. During the current COVID-19 pandemic, recent advances in mRNA engineering have proven paramount in the rapid design and production of effective vaccines. Comparatively, however, the development of new adjuvants capable of enhancing vaccine efficacy has been lagging. Despite massive improvements in our understanding of immunology, fewer than ten adjuvants have been approved for human use in the century since the discovery of the first adjuvant. Modern adjuvants can improve vaccines against future pathogens by reducing cost, improving antigen immunogenicity, and increasing antigen stability. In this perspective, we survey the current state of adjuvant use, highlight potentially impactful preclinical adjuvants, and propose new measures to accelerate adjuvant safety testing and technology sharing to enable the use of "off-the-shelf" adjuvant platforms for rapid vaccine testing and deployment in the face of future pandemics.
Collapse
Affiliation(s)
| | - Kevin J. McHugh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA;
| |
Collapse
|
43
|
Tang M, Li S, Wei L, Hou Z, Qu J, Li L. Do Engineered Nanomaterials Affect Immune Responses by Interacting With Gut Microbiota? Front Immunol 2021; 12:684605. [PMID: 34594323 PMCID: PMC8476765 DOI: 10.3389/fimmu.2021.684605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/26/2021] [Indexed: 12/30/2022] Open
Abstract
Engineered nanomaterials (ENMs) have been widely exploited in several industrial domains as well as our daily life, raising concern over their potential adverse effects. While in general ENMs do not seem to have detrimental effects on immunity or induce severe inflammation, their indirect effects on immunity are less known. In particular, since the gut microbiota has been tightly associated with human health and immunity, it is possible that ingested ENMs could affect intestinal immunity indirectly by modulating the microbial community composition and functions. In this perspective, we provide a few pieces of evidence and discuss a possible link connecting ENM exposure, gut microbiota and host immune response. Some experimental works suggest that excessive exposure to ENMs could reshape the gut microbiota, thereby modulating the epithelium integrity and the inflammatory state in the intestine. Within such microenvironment, numerous microbiota-derived components, including but not limited to SCFAs and LPS, may serve as important effectors responsible of the ENM effect on intestinal immunity. Therefore, the gut microbiota is implicated as a crucial regulator of the intestinal immunity upon ENM exposure. This calls for including gut microbiota analysis within future work to assess ENM biocompatibility and immunosafety. This also calls for refinement of future studies that should be designed more elaborately and realistically to mimic the human exposure situation.
Collapse
Affiliation(s)
- Mingxing Tang
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Shuo Li
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Lan Wei
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,School of Biomedical Science and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Zhaohua Hou
- Department of Surgery, Sloan Kettering Institute Z427-D, Mortimer B. Zuckerman Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jing Qu
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
44
|
Gonzalez I, Araya P, Schneider I, Lindner C, Rojas A. Pattern recognition receptors and their roles in the host response to Helicobacter pylori infection. Future Microbiol 2021; 16:1229-1238. [PMID: 34615380 DOI: 10.2217/fmb-2021-0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (H. pylori) infection is highly prevalent, affecting 4.4 billion people globally. This pathogen is a risk factor in the pathogenesis of more than 75% of worldwide cases of gastric cancer. Pattern recognition receptors are essential in the innate immune response to H. pylori infection. They recognize conserved pathogen structures and myriad alarmins released by host cells in response to microbial components, cytokines or cellular stress, thus triggering a robust proinflammatory response, which is crucial in H. pylori-induced gastric carcinogenesis. In this review, we intend to highlight the main pattern recognition receptors involved in the recognition and host response to H. pylori, as well as the main structures recognized and the subsequent inflammatory response.
Collapse
Affiliation(s)
- Ileana Gonzalez
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, 3460000, Chile
| | - Paulina Araya
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, 3460000, Chile
| | - Ivan Schneider
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, 3460000, Chile
| | - Cristian Lindner
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, 3460000, Chile
| | - Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, 3460000, Chile
| |
Collapse
|
45
|
Bacterial Flagellar Filament: A Supramolecular Multifunctional Nanostructure. Int J Mol Sci 2021; 22:ijms22147521. [PMID: 34299141 PMCID: PMC8306008 DOI: 10.3390/ijms22147521] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/06/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023] Open
Abstract
The bacterial flagellum is a complex and dynamic nanomachine that propels bacteria through liquids. It consists of a basal body, a hook, and a long filament. The flagellar filament is composed of thousands of copies of the protein flagellin (FliC) arranged helically and ending with a filament cap composed of an oligomer of the protein FliD. The overall structure of the filament core is preserved across bacterial species, while the outer domains exhibit high variability, and in some cases are even completely absent. Flagellar assembly is a complex and energetically costly process triggered by environmental stimuli and, accordingly, highly regulated on transcriptional, translational and post-translational levels. Apart from its role in locomotion, the filament is critically important in several other aspects of bacterial survival, reproduction and pathogenicity, such as adhesion to surfaces, secretion of virulence factors and formation of biofilms. Additionally, due to its ability to provoke potent immune responses, flagellins have a role as adjuvants in vaccine development. In this review, we summarize the latest knowledge on the structure of flagellins, capping proteins and filaments, as well as their regulation and role during the colonization and infection of the host.
Collapse
|
46
|
Xia P, Wu Y, Lian S, Yan L, Meng X, Duan Q, Zhu G. Research progress on Toll-like receptor signal transduction and its roles in antimicrobial immune responses. Appl Microbiol Biotechnol 2021; 105:5341-5355. [PMID: 34180006 PMCID: PMC8236385 DOI: 10.1007/s00253-021-11406-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
When microorganisms invade a host, the innate immune system first recognizes the pathogen-associated molecular patterns of these microorganisms through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are known transmembrane PRRs existing in both invertebrates and vertebrates. Upon ligand recognition, TLRs initiate a cascade of signaling events; promote the pro-inflammatory cytokine, type I interferon, and chemokine expression; and play an essential role in the modulation of the host's innate and adaptive immunity. Therefore, it is of great significance to improve our understanding of antimicrobial immune responses by studying the role of TLRs and their signal molecules in the host's defense against invading microbes. This paper aims to summarize the specificity of TLRs in recognition of conserved microbial components, such as lipoprotein, lipopolysaccharide, flagella, endosomal nucleic acids, and other bioactive metabolites derived from microbes. This set of interactions helps to elucidate the immunomodulatory effect of TLRs and the signal transduction changes involved in the infectious process and provide a novel therapeutic strategy to combat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Xia Meng
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Qiangde Duan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| |
Collapse
|
47
|
Razim A, Pacyga K, Naporowski P, Martynowski D, Szuba A, Gamian A, Górska S. Identification of linear epitopes on the flagellar proteins of Clostridioides difficile. Sci Rep 2021; 11:9940. [PMID: 33976336 PMCID: PMC8113543 DOI: 10.1038/s41598-021-89488-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
Clostridioides difficile (C. difficile) is an opportunistic anaerobic bacterium that causes severe diseases of the digestive tract of humans and animals. One of the possible methods of preventing C. difficile infection is to develop a vaccine. The most promising candidates for vaccine antigens are the proteins involved in the adhesion phenomena. Among them, the FliC and FliD are considered to be suitable candidates. In this paper, the FliC and FliD protein polypeptide epitopes were mapped in silico and by using PEPSCAN procedure. We identified four promising epitopes: 117QRMRTLS123, 205MSKAG209 of FliC and 226NKVAS230, 306TTKKPKD312 of FliD protein. We showed that 117QRMRTLS123 sequence is not only located in TLR5-binding and activating region, as previously shown, but forms an epitope recognized by C. difficile-infected patients' antibodies. 205MSKAG209 is a C. difficile-unique, immunogenic sequence that forms an exposed epitope on the polymerized flagella structure which makes it a suitable vaccine antigen. 226NKVAS230 and 306TTKKPKD312 are well exposed and possess potential protective properties according to VaxiJen analysis. Our results open the possibility to use these epitopes as suitable anti-C. difficile vaccine antigens.
Collapse
Affiliation(s)
- A Razim
- Laboratory of Immunobiology of Microbiome, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland.
| | - K Pacyga
- Laboratory of Immunobiology of Microbiome, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland
| | - P Naporowski
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland
| | - D Martynowski
- Laboratory of Genomics and Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland
| | - A Szuba
- Division of Angiology, Wroclaw Medical University, Wroclaw, Poland.,Department of Internal Medicine, 4th Military Hospital in Wroclaw, Wroclaw, Poland
| | - A Gamian
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland
| | - S Górska
- Laboratory of Immunobiology of Microbiome, Hirszfeld Institute of Immunology and Experimental Therapy, PAS, Wroclaw, Poland
| |
Collapse
|
48
|
Toll-like Receptor 5 Activation by the CagY Repeat Domains of Helicobacter pylori. Cell Rep 2021; 32:108159. [PMID: 32937132 DOI: 10.1016/j.celrep.2020.108159] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/01/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (Hp) is an important human pathogen associated with gastric inflammation and neoplasia. It is commonly believed that this bacterium avoids major immune recognition by Toll-like receptors (TLRs) because of low intrinsic activity of its flagellin and lipopolysaccharides (LPS). In particular, TLR5 specifically detects flagellins in various bacterial pathogens, while Hp evolved mutations in flagellin to evade detection through TLR5. Cancerogenic Hp strains encode a type IV secretion system (T4SS). The T4SS core component and pilus-associated protein CagY, a large VirB10 ortholog, drives effector molecule translocation. Here, we identify CagY as a flagellin-independent TLR5 agonist. We detect five TLR5 interaction sites, promoting binding of CagY-positive Hp to TLR5-expressing cells, TLR5 stimulation, and intracellular signal transduction. Consequently, CagY constitutes a remarkable VirB10 member detected by TLR5, driving crucial innate immune responses by this human pathogen.
Collapse
|
49
|
Herrera LRM. Reverse Vaccinology Approach in Constructing a Multi-Epitope Vaccine Against Cancer-Testis Antigens Expressed in Non-Small Cell Lung Cancer. Asian Pac J Cancer Prev 2021; 22:1495-1506. [PMID: 34048178 PMCID: PMC8408400 DOI: 10.31557/apjcp.2021.22.5.1495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Indexed: 12/26/2022] Open
Abstract
Background: The 5-year survival rate of non-small cell lung cancer (NSCLC) patients has not significantly improved despite advancements in the currently applied treatments. Thus, efforts are put forth in developing novel immunotherapeutic agents targeting cancer-testis antigens (CTA) in NSCLC. This work utilized reverse vaccinology approach in designing a novel multi-epitope vaccine targeting melanoma-associated antigen 3 (MAGEA3), MAGEA4, New York esophageal squamous cell carcinoma-1 (NY-ESO-1), and Kita-Kyushu lung cancer antigen 1 (KK-LC1), being the most frequently expressed CTAs in NSCLC. Methods: Epitopes were mapped from the sequences of CTAs. The population coverage (PC) of identified CD4+ and CD8+ epitopes were estimated. Candidate linear B cell (BL), CD4+, and CD8+ epitopes were adjoined in a multi-epitope construct (Mvax) with flagellin domain as an adjuvant. Antigenicity, and cross-reactivity of Mvax were examined. The tertiary structure of Mvax was modelled, and validated. All epitopes included in the vaccine were docked with their human leukocyte antigen (HLA) binders. The immunogenicity of epitopes in Mvax was validated through molecular dynamics analysis. Results: Mvax contains 22 epitopes from MAGEA3, MAGEA4, NY-ESO-1, and KK-LC1. It is classified as antigenic, non-allergen, non-toxic, and possesses physicochemical stability. Epitopes have no significant hits with other human proteins, except for 2 other CTAs frequently expressed in NSCLC. The stretch of BL epitopes in Mvax confers flexibility, and accessibility emphasizing its antigenicity. The tertiary structure analysis showed that Mvax model has good structural quality. All epitopes included in the vaccine are highly immunogenic as indicated by favorable binding affinity, low binding energy, and acceptable root-mean-square deviation (RMSD). CD4+ and CD8+ epitopes have global PC of 81.81%, and 84.15%, respectively. Conclusion: Overall, in silico evaluations show that Mvax is a potential immunotherapeutic agent against NSCLC.
Collapse
Affiliation(s)
- Leana Rich M Herrera
- Department of Physical Sciences, College of Science, Polytechnic University of the Philippines, Manila City, Philippines
| |
Collapse
|
50
|
Elmi A, Nasher F, Dorrell N, Wren B, Gundogdu O. Revisiting Campylobacter jejuni Virulence and Fitness Factors: Role in Sensing, Adapting, and Competing. Front Cell Infect Microbiol 2021; 10:607704. [PMID: 33614526 PMCID: PMC7887314 DOI: 10.3389/fcimb.2020.607704] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial foodborne gastroenteritis world wide and represents a major public health concern. Over the past two decades, significant progress in functional genomics, proteomics, enzymatic-based virulence profiling (EBVP), and the cellular biology of C. jejuni have improved our basic understanding of this important pathogen. We review key advances in our understanding of the multitude of emerging virulence factors that influence the outcome of C. jejuni–mediated infections. We highlight, the spatial and temporal dynamics of factors that promote C. jejuni to sense, adapt and survive in multiple hosts. Finally, we propose cohesive research directions to obtain a comprehensive understanding of C. jejuni virulence mechanisms.
Collapse
Affiliation(s)
- Abdi Elmi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fauzy Nasher
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nick Dorrell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Brendan Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|