Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 14.
Article in English | MEDLINE | ID: mdl-38256103

ABSTRACT

S100A8, S100A9, and S100A12 proteins are important members of the S100 protein family, act primarily as congenital immunomodulators, and are closely related to the occurrence of infectious diseases. There have been few reports on the functional properties of S100A8, S100A9, and S100A12 proteins in swine, but it is certain that porcine S100A8, S100A9, and S100A12 proteins are highly expressed in diseased swine. To address the current lack of reliable and timely detection tools for these three proteins, we generated monoclonal antibodies specific to the porcine S100A8, S100A9, and S100A12 proteins using hybridoma technology. The results of serum sample testing showed that the above monoclonal antibodies specifically recognize the proteins S100A8, S100A9, and S100A12 in the serum and were able to evaluate the content change of these proteins during the infection process. This provides the basis for the use of porcine S100A8, S100A9, and S100A12 in the surveillance and diagnosis of swine diseases and laid a foundation for further understanding their roles in infection, immunity, and inflammation, as well as their potential applications in preventing or treating gastrointestinal tract or inflammatory diseases in swine.


Subject(s)
Antibodies, Monoclonal , S100A12 Protein , Swine , Animals , Hybridomas , Calgranulin A , Calgranulin B , Technology
2.
Immunology ; 171(3): 365-376, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38013255

ABSTRACT

S100 proteins are small proteins that are only expressed in vertebrates. They are widely expressed in many different cell types and are involved in the regulation of calcium homeostasis, glucose metabolism, cell proliferation, apoptosis, inflammation and tumorigenesis. As members of the S100 protein subfamily of myeloid-related proteins, S100A8, S100A9 and S100A12 play a crucial role in resisting microbial infection and maintaining immune homeostasis. These proteins chelate the necessary metal nutrients of pathogens invading the host by means of 'nutritional immunity' and directly inhibit the growth of pathogens in the host. They interact with receptors on the cell surface to initiate inflammatory signal transduction, induce cytokine expression and participate in the inflammatory response and immune regulation. Furthermore, the increased content of these proteins during the pathological process makes them useful as disease markers for screening and detecting related diseases. This article summarizes the structure and function of the proteins S100A8, S100A9 and S100A12 and lays the foundation for further understanding their roles in infection, immunity and inflammation, as well as their potential applications in the prevention and treatment of infectious diseases.


Subject(s)
Inflammation , S100A12 Protein , Animals , Humans , Calgranulin B , Calgranulin A/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism
3.
Acta Trop ; 249: 107082, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38008371

ABSTRACT

Assessing the risk of malaria local transmission and re-introduction is crucial for the preparation and implementation of an effective elimination campaign and the prevention of malaria re-introduction in China. Therefore, this review aims to evaluate the risk factors for malaria local transmission and re-introduction in China over the period of pre-elimination to elimination. Data were obtained from six databases searched for studies that assessed malaria local transmission risk before malaria elimination and re-introduction risk after the achievement of malaria elimination in China since the launch of the NMEP in 2010, employing the keywords "malaria" AND ("transmission" OR "re-introduction") and their synonyms. A total of 8,124 articles were screened and 53 articles describing 55 malaria risk assessment models in China from 2010 to 2023, including 40 models assessing malaria local transmission risk (72.7%) and 15 models assessing malaria re-introduction risk (27.3%). Factors incorporated in the 55 models were extracted and classified into six categories, including environmental and meteorological factors (39/55, 70.9%), historical epidemiology (35/55, 63.6%), vectorial factors (32/55, 58.2%), socio-demographic information (15/26, 53.8%), factors related to surveillance and response capacity (18/55, 32.7%), and population migration aspects (13/55, 23.6%). Environmental and meteorological factors as well as vectorial factors were most commonly incorporated in models assessing malaria local transmission risk (29/40, 72.5% and 21/40, 52.5%) and re-introduction risk (10/15, 66.7% and 11/15, 73.3%). Factors related to surveillance and response capacity and population migration were also important in malaria re-introduction risk models (9/15, 60%, and 6/15, 40.0%). A total of 18 models (18/55, 32.7%) reported the modeling performance. Only six models were validated internally and five models were validated externally. Of 53 incorporated studies, 45 studies had a quality assessment score of seven and above. Environmental and meteorological factors as well as vectorial factors play a significant role in malaria local transmission and re-introduction risk assessment. The factors related to surveillance and response capacity and population migration are more important in assessing malaria re-introduction risk. The internal and external validation of the existing models needs to be strengthened in future studies.


Subject(s)
Malaria , Humans , Malaria/epidemiology , Malaria/prevention & control , China/epidemiology , Risk Factors , Risk Assessment , Meteorological Concepts
4.
Microorganisms ; 11(1)2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36677506

ABSTRACT

RyhB-1 and RyhB-2 are small non-coding RNAs in Salmonella that act as regulators of iron homeostasis by sensing the environmental iron concentration. Expressions of RyhB paralogs from Salmonella Typhimurium are increased within microphages. RyhB paralogs restrain the growth of S. Typhimurium in RAW264.7 macrophages by modulating the expression of Salmonella pathogenicity island 1 (SPI-1) genes sicA and rtsB. However, little is known about the regulatory role of RyhBs and their virulence-associated targets in Salmonella Enteritidis. We studied candidate targets of RyhB paralogs via RNA-Seq in conditions of iron limitation and hypoxia. RyhB paralogs were expressed when the S. Enteritidis strain CMCC(B)50336 (SE50336) interacted with the chicken macrophage line HD11. We analyzed gene expression associated with Salmonella survival and replication in macrophages in wild-type strain SE50336 and the RyhB deletion mutants after co-incubation with HD11 and screened out targets regulated by RyhBs. The expressions of both RyhB-1 and RyhB-2 were increased after co-incubation with HD11 for 8 h and several survival-associated genes within macrophages, such as ssaI, sseA, pagC, sodC, mgtC, yaeB, pocR, and hns, were upregulated in the ryhB-1 deletion mutant. Specifically, ssaI, the type-three secretion system 2 (T3SS-2) effector encoded by SPI-2, which promoted the survival of Salmonella in macrophages, was upregulated more than 3-fold in the ryhB-1 deletion mutant. We confirmed that both RyhB-1 and RyhB-2 downregulated the expression of ssaI to repress its mRNA translation by directly interacting with its coding sequence (CDS) region via an incomplete complementary base-pairing mechanism. The SPI-2 gene sseA was indirectly modulated by RyhB-1. The survival assays in macrophages showed that the ability of intracellular survival of ryhB-1 and/or ryhB-2 deletion mutants in HD11 was higher than that of the wild-type strain. These results indicate that RyhB paralogs downregulate survival-related virulence factors and attenuate the survival of S. Enteritidis inside chicken macrophage HD11.

5.
Int J Mol Sci ; 25(1)2023 Dec 24.
Article in English | MEDLINE | ID: mdl-38203452

ABSTRACT

Bacteria have existed on Earth for billions of years, exhibiting ubiquity and involvement in various biological activities. To ensure survival, bacteria usually release and secrete effector proteins to acquire nutrients and compete with other microorganisms for living space during long-term evolution. Consequently, bacteria have developed a range of secretion systems, which are complex macromolecular transport machines responsible for transporting proteins across the bacterial cell membranes. Among them, one particular secretion system that stands out from the rest is the type V secretion system (T5SS), known as the "autotransporter". Bacterial activities mediated by T5SS include adherence to host cells or the extracellular matrix, invasion of host cells, immune evasion and serum resistance, contact-dependent growth inhibition, cytotoxicity, intracellular flow, protease activity, autoaggregation, and biofilm formation. In a bacterial body, it is not enough to rely on T5SS alone; in most cases, T5SS cooperates with other secretion systems to carry out bacterial life activities, but regardless of how good the relationship is, there is friction between the secretion systems. T5SS and T1SS/T2SS/T3SS/T6SS all play a synergistic role in the pathogenic processes of bacteria, such as nutrient acquisition, pathogenicity enhancement, and immune modulation, but T5SS indirectly inhibits the function of T4SS. This could be considered a love-hate relationship between secretion systems. This paper uses the systematic literature review methodology to review 117 journal articles published within the period from 1995 to 2024, which are all available from the PubMed, Web of Science, and Scopus databases and aim to elucidate the link between T5SS and other secretion systems, providing clues for future prevention and control of bacterial diseases.


Subject(s)
Bacteria , Type V Secretion Systems , Bodily Secretions , Cell Aggregation , Cell Membrane
6.
Int J Mol Sci ; 23(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36292986

ABSTRACT

G protein-coupled receptor 39 (GPR39) is a zinc-sensing receptor (ZnR) that can sense changes in extracellular Zn2+, mediate Zn2+ signal transmission, and participate in the regulation of numerous physiological activities in living organisms. For example, GPR39 activates the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) and phosphatidylinositol3-kinase/protein kinase B (PI3K/AKT) signaling pathways upon Zn2+ stimulation, enhances the proliferation and differentiation of colonic cells, and regulates ion transport, as well as exerting other functions. In recent years, with the increased attention to animal gut health issues and the intensive research on GPR39, GPR39 has become a potential target for regulating animal intestinal health. On the one hand, GPR39 is involved in regulating ion transport in the animal intestine, mediating the Cl- efflux by activating the K+/Cl- synergistic protein transporter, and relieving diarrhea symptoms. On the other hand, GPR39 can maintain the homeostasis of the animal intestine, promoting pH restoration in colonic cells, regulating gastric acid secretion, and facilitating nutrient absorption. In addition, GPR39 can affect the expression of tight junction proteins in intestinal epithelial cells, improving the barrier function of the animal intestinal mucosa, and maintaining the integrity of the intestine. This review summarizes the structure and signaling transduction processes involving GPR39 and the effect of GPR39 on the regulation of intestinal health in animals, with the aim of further highlighting the role of GPR39 in regulating animal intestinal health and providing new directions and ideas for studying the prevention and treatment of animal intestinal diseases.


Subject(s)
Proto-Oncogene Proteins c-akt , Zinc , Animals , Zinc/metabolism , Phosphatidylinositol 3-Kinases , Receptors, G-Protein-Coupled/metabolism , Tight Junction Proteins , Extracellular Signal-Regulated MAP Kinases
7.
Biology (Basel) ; 11(9)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36138763

ABSTRACT

Salmonella species infect hosts by entering phagocytic and non-phagocytic cells, causing diverse disease symptoms, such as fever, gastroenteritis, and even death. Therefore, Salmonella has attracted much attention. Many factors are involved in pathogenesis, for example, the capsule, enterotoxins, Salmonella pathogenicity islands (SPIs), and corresponding regulators. These factors are all traditional proteins associated with virulence and regulation. Recently, small non-coding RNAs (sRNAs) have also been reported to function as critical regulators. Salmonella has become a model organism for studying sRNAs. sRNAs regulate gene expression by imperfect base-pairing with targets at the post-transcriptional level. sRNAs are involved in diverse biological processes, such as virulence, substance metabolism, and adaptation to stress environments. Although some studies have reported the crucial roles of sRNAs in regulating host-pathogen interactions, the function of sRNAs in host-Salmonella interactions has rarely been reviewed. Here, we review the functions of sRNAs during the infection of host cells by Salmonella, aiming to deepen our understanding of sRNA functions and the pathogenic mechanism of Salmonella.

8.
PLoS One ; 17(7): e0269102, 2022.
Article in English | MEDLINE | ID: mdl-35900973

ABSTRACT

Avian pathogenic Escherichia coli (APEC), which has potential zoonotic risk, can cause severe systemic infections such as septicemia and meningitis in poultry. Colibactin is a hybrid non-ribosomal peptide/polyketide secondary metabolite produced by bacteria, which induces double-strand DNA breaks and chromosome instability in eukaryotic cells. ClbA is a 4'-phosphopantetheinyl transferase (PPTase) that is essential for colibactin and plays a role in siderophore synthesis. However, whether ClbA is associated with meningitis development in APEC is unclear. In this study, we abolished the clbA gene in the APEC XM strain, investigated the effect of clbA on colibactin synthesis and evaluated the pathogenic capacity of colibactin on meningitis development. Deletion of clbA reduced DNA damage to cells and hindered the normal synthesis of colibactin. Compared with the mice infected by wild-type APEC XM, the clbA deletion mutant infected mice had significant reduction in a series of characteristics associated with meningitis including clinical symptoms, bacterial loads of blood and brain, disruption of the blood brain barrier and the expression of inflammatory factors in the brain tissue. Complementation of ClbA recovered some APEC XM virulence. We conclude that ClbA is obligatory for the synthesis of colibactin and is responsible for the development of meningitis in mice infected by APEC.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Meningitis , Poultry Diseases , Animals , Bacterial Proteins , Birds/metabolism , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Mice , Poultry Diseases/microbiology , Transferases (Other Substituted Phosphate Groups) , Virulence/genetics , Virulence Factors/metabolism
9.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216425

ABSTRACT

Bacteria and viruses are both important pathogens causing intestinal infections, and studies on their pathogenic mechanisms tend to focus on one pathogen alone. However, bacterial and viral co-infections occur frequently in clinical settings, and infection by one pathogen can affect the severity of infection by another pathogen, either directly or indirectly. The presence of synergistic or antagonistic effects of two pathogens in co-infection can affect disease progression to varying degrees. The triad of bacterial-viral-gut interactions involves multiple aspects of inflammatory and immune signaling, neuroimmunity, nutritional immunity, and the gut microbiome. In this review, we discussed the different scenarios triggered by different orders of bacterial and viral infections in the gut and summarized the possible mechanisms of synergy or antagonism involved in their co-infection. We also explored the regulatory mechanisms of bacterial-viral co-infection at the host intestinal immune interface from multiple perspectives.


Subject(s)
Bacterial Infections/immunology , Coinfection/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Virus Diseases/immunology , Animals , Coinfection/microbiology , Coinfection/virology , Humans , Intestinal Mucosa/virology
10.
Virulence ; 12(1): 2382-2399, 2021 12.
Article in English | MEDLINE | ID: mdl-34529552

ABSTRACT

Colibactin is synthesized by a 54-kb genomic island, leads to toxicity in eukaryotic cells, and plays a vital role in many diseases, including neonatal sepsis and meningitis. Avian pathogenic Escherichia coli (APEC) is speculated to be an armory of extraintestinal pathogenic Escherichia coli and can be a potential zoonotic bacterium that threatens human and animal health. In this study, the APEC XM meningitis mouse model was successfully established to investigate the effect of colibactin in in vivo infection. The clbH-deletion mutant strain induced lower γ-H2AX expression, no megalocytosis, and no cell cycle arrest in bEnd.3 cells, which showed that the deletion of clbH decreased the production of colibactin in the APEC XM strain. The deletion of clbH did not affect the APEC XM strain's ability of adhering to and invading bEnd.3 cells. In vitro, the non-colibactin-producing strain displayed significantly lower serum resistance and it also induced a lower level of cytokine mRNA and few disruptions of tight junction proteins in infected bEnd.3 cells. Meningitis did not occur in APEC ΔclbH-infected mice in vivo, who showed fewer clinical symptoms and fewer lesions on radiological and histopathological analyses. Compared with the APEX XM strain, APEC ΔclbH induced lower bacterial colonization in tissues, lower mRNA expression of cytokines in brain tissues, and slight destruction of the brain blood barrier. These results indicate that clbH is a necessary component for the synthesis of genotoxic colibactin, and colibactin is related to the development of meningitis induced by APEC XM.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Meningitis , Animals , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mice , Peptides , Polyketides , RNA, Messenger
11.
Toxins (Basel) ; 13(8)2021 08 06.
Article in English | MEDLINE | ID: mdl-34437417

ABSTRACT

Colibactin is a complex secondary metabolite that leads to genotoxicity that interferes with the eukaryotic cell cycle. It plays an important role in many diseases, including neonatal mouse sepsis and meningitis. Avian pathogenic Escherichia coli (APEC) is responsible for several diseases in the poultry industry and may threaten human health due to its potential zoonosis. In this study, we confirmed that clbG was necessary for the APEC XM strain to produce colibactin. The deletion of clbG on APEC XM contributed to lowered γH2AX expression, no megalocytosis, and no cell cycle arrest in vitro. None of the 4-week Institute of Cancer Research mice infected with the APEC XM ΔclbG contracted meningitis or displayed weakened clinical symptoms. Fewer histopathological lesions were observed in the APEC XM ΔclbG group. The bacterial colonization of tissues and the relative expression of cytokines (IL-1ß, IL-6, and TNF-α) in the brains decreased significantly in the APEC XM ΔclbG group compared to those in the APEC XM group. The tight junction proteins (claudin-5, occludin, and ZO-1) were not significantly destroyed in APEC XM ΔclbG group in vivo and in vitro. In conclusion, clbG is necessary for the synthesis of the genotoxin colibactin and affects the development of APEC meningitis in mice.


Subject(s)
Escherichia coli Infections , Peptides/toxicity , Polyketides/toxicity , Animals , Brain/metabolism , Brain/pathology , Cell Line , Cytokines/genetics , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli Infections/veterinary , Female , Male , Mice, Inbred ICR , Peptides/genetics , Peptides/metabolism , Polyketides/metabolism , Poultry Diseases , Tight Junction Proteins/metabolism
12.
Arch Microbiol ; 203(7): 4221-4231, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34091701

ABSTRACT

The Escherichia coli (E. coli) nirC gene encodes a nitrite transporter, which involved in transporting toxic nitrite (NO2-) from the environment into the bacteria. Although the deletion of nirC gene could cause changes in motility, adhesion in the previous study, and the virulence involved in the specified mechanism for pathogenic E. coli remains to be known. In the present work, we aimed to evaluate the role of NirC in a serotype O2:K1:H7 avian pathogenic Escherichia coli (APEC) strain. For this purpose, we generated a NirC-deficient mutant of APEC XM strain and examined its biological characteristics. The nirC gene deletion mutant enhanced ability of motility, decreased in biofilm formation, and it markedly reduced ability to adhere mouse brain microvascular endothelial cell b.End3 cells. For understanding its mechanism, sequentially we detected and found the stress regulator rpoS and its downstream genes csrA were up-regulated in NirC-deficient mutant while diguanylate cyclase gene dgcT was down-regulated. By high-performance liquid chromatography (HPLC) experiment, we demonstrated the concentration of intracellular 3',5'-cyclic diguanosine monophosphate (c-di-GMP) significantly decrease in nirC gene deletion mutant. Taken data together, we may make a conclusion with a possible signal pathway clue, due to NirC mutation, environmental NO2- accumulation leads to nitrite stress and inactivates c-di-GMP synthesis by stimulating the stress regulator RpoS, resulting in changes of biological characteristics.


Subject(s)
Anion Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Animals , Anion Transport Proteins/genetics , Bacterial Adhesion/genetics , Biofilms , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Mice , Mutation , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Virulence/genetics
13.
J Vis Exp ; (171)2021 05 18.
Article in English | MEDLINE | ID: mdl-34096912

ABSTRACT

Porcine aminopeptidase N (APN), a membrane-bound metallopeptidase abundantly present in small intestinal mucosa, can initiate a mucosal immune response without any interference such as low protein expression, enzyme inactivity, or structural changes. This makes APN an attractive candidate in the development of vaccines that selectively target the mucosal epithelium. Previous studies have shown that APN is a receptor protein for both enterotoxigenic Escherichia coli (E. coli) F4 and transmissible gastroenteritis virus. Thus, APN shows promise in the development of antibody-drug conjugates or novel vaccines based on APN-specific antibodies. In this study, we compared production of APN-specific monoclonal antibodies (mAbs) using traditional hybridoma technology and recombinant antibody expression method. We also established a stably transfected Chinese hamster ovary (CHO) cell line using pIRES2-ZSGreen1-rAbs-APN and an E. coli expression BL21(DE3) strain harboring the pET28a (+)-rAbs-APN vector. The results show that antibodies expressed in pIRES2-ZSGreen1-rAbs-APN-CHO cells and mAbs produced using hybridomas could recognize and bind to the APN protein. This provides the basis for further elucidation of the APN receptor function for the development of therapeutics targeting different APN-specific epitopes.


Subject(s)
Antibodies, Monoclonal , CD13 Antigens , Intestinal Mucosa , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Cricetulus , Epithelium , Escherichia coli , Intestinal Mucosa/immunology , Swine
14.
Mol Immunol ; 136: 45-54, 2021 08.
Article in English | MEDLINE | ID: mdl-34082258

ABSTRACT

Toll-like receptor 8 (TLR8), as an important innate immune receptor, can recognize specific ligands, activate intracellular signaling and produce an inflammatory response to kill and eliminate pathogenic microorganisms. Recent studies have resolved the crystal structure of human TLR8 (hTLR8) and two types of ligand binding sites were identified. Among the conserved binding site 1 of hTLR8, the residues interacting with imidazoquinoline derivatives (IQDs) were determined. We previously showed that porcine TLR8 (pTLR8) exhibited species specificity for recognition of the hTLR7 agonist imiquimod (R837). Given the species specificity, the pTLR8 residues interacting with IQDs may be different from hTLR8 counterparts. The present study was aimed to identify the pTLR8 residues interacting with small molecular IQDs. Via molecular docking, the pTLR8 residues interacting with R837 and R848 were predicted. The corresponding mutants were tested for pTLR8 signaling in response to IQDs R837, R848 and CL075, and the results showed that five of nine predicted residues (Y336, K341, K342, F395 and G562) are critical for pTLR8 signaling and these residues are partially different from those reported in hTLR8. Further, we found that the pTLR8 GQKNG motif corresponding to hTLR8 RQSYA exhibited disparity to CL075 stimulation. Our study thus reveals fine TLR8 species specificity which deepens the understanding of TLR8 activation mechanism.


Subject(s)
Imidazoles/metabolism , Quinolines/metabolism , Toll-Like Receptor 8/metabolism , Amino Acid Sequence/genetics , Animals , Binding Sites/physiology , Cell Line , HEK293 Cells , Humans , Imiquimod/pharmacology , Immunity, Innate/immunology , Molecular Conformation , Molecular Docking Simulation , Protein Domains/immunology , Signal Transduction/genetics , Species Specificity , Swine , Toll-Like Receptor 8/genetics
15.
Appl Microbiol Biotechnol ; 105(13): 5341-5355, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34180006

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.


Subject(s)
Anti-Infective Agents , Immunity, Innate , Adaptive Immunity , Animals , Signal Transduction , Toll-Like Receptors
16.
Appl Microbiol Biotechnol ; 105(13): 5631-5641, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34155530

ABSTRACT

Salmonella Enteritidis (SE) causes both horizontal and vertical transmission of diseases in poultry industry and is also one of the main causes of human food poisoning. Sequence analysis of the sef operon of poultry-derived Salmonella serotypes showed the presence of an entire sef operon in SE, whereas only sef pseudogenes were found in Salmonella Gallinarum and Salmonella Pullorum. Subsequently, the sef operon of SE was cloned into the pBR322 plasmid and expressed in a modified Escherichia coli strain SE5000. sef operon expression was demonstrated using sodium dodecyl sulfate-polyacrylamide gel electrophoresis, western blot, agglutination assay, and transmission electron microscopy. The results showed that SE5000+Sef, but not SE5000+pBR322, could specifically react with SE-positive chicken serum in an agglutination assay, which could be clearly visualized by the naked eye within less than 2 min. In contrast, SE5000+Sef could not be recognized in Salmonella Gallinarum- and Salmonella Pullorum-positive chicken sera. Next, taking advantage of the exclusive presence of an entire sef operon in SE, we set up an agglutination-based detection system to monitor the dynamics of Sef-targeted antibody from SE-infected chicks for 47 days. Using the proposed detection method, SE was readily detectable starting from 2 weeks post-infection. Finally, we compared the proposed SE5000+Sef-based detection system with commercially available agglutination antigen using the classical bacterial isolation and identification procedure as reference. The results showed that the SE5000+Sef system was more consistent with the results of bacterial isolation and identification with almost 100% accuracy. We established a simple, sensitive, and cheap agglutination method for rapid and specific detection of SE-infected chickens, which can facilitate epidemiological investigation and eradication of SE infections. KEY POINTS: • Only the Salmonella Enteritidis serotype expressed Sef fimbriae in chicken infected with SE. • A rapid, large-scale method of detection by the naked eye of detection of SE-infected chicken is presented.


Subject(s)
Poultry Diseases , Salmonella Infections, Animal , Animals , Chickens , Fimbriae, Bacterial , Humans , Operon , Poultry Diseases/diagnosis , Salmonella enteritidis/genetics
17.
Vet Res ; 52(1): 44, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33691799

ABSTRACT

Small non-coding RNA RyhB is a key regulator of iron homeostasis in bacteria by sensing iron availability in the environment. Although RyhB is known to influence bacterial virulence by interacting with iron metabolism related regulators, its interaction with virulence genes, especially the Type III secretion system (T3SS), has not been reported. Here, we demonstrate that two RyhB paralogs of Salmonella enterica serovar Enteritidis upregulate Type III secretion system (T3SS) effectors, and consequently affect Salmonella invasion into intestinal epithelial cells. Specifically, we found that RyhB-1 modulate Salmonella response to stress condition of iron deficiency and hypoxia, and stress in simulated intestinal environment (SIE). Under SIE culture conditions, both RyhB-1 and RyhB-2 are drastically induced and directly upregulate the expression of T3SS effector gene sipA by interacting with its 5' untranslated region (5' UTR) via an incomplete base-pairing mechanism. In addition, the RyhB paralogs upregulate the expression of T3SS effector gene sopE. By regulating the invasion-related genes, RyhBs in turn affect the ability of S. Enteritidis to adhere to and invade into intestinal epithelial cells. Our findings provide evidence that RyhBs function as critical virulence factors by directly regulating virulence-related gene expression. Thus, inhibition of RyhBs may be a potential strategy to attenuate Salmonella.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Microfilament Proteins/genetics , Salmonella enteritidis/physiology , Salmonella enteritidis/pathogenicity , Virulence Factors/genetics , Animals , Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Genes, Bacterial/genetics , Intestines/microbiology , Microfilament Proteins/metabolism , Salmonella Infections, Animal/microbiology , Up-Regulation , Virulence
18.
Vet Res ; 52(1): 39, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33663613

ABSTRACT

Zinc (Zn) is an essential trace element in living organisms and plays a vital role in the regulation of both microbial virulence and host immune responses. A growing number of studies have shown that zinc deficiency or the internal Zn concentration does not meet the needs of animals and microbes, leading to an imbalance in zinc homeostasis and intracellular signalling pathway dysregulation. Competition for zinc ions (Zn2+) between microbes and the host exists in the use of Zn2+ to maintain cell structure and physiological functions. It also affects the interplay between microbial virulence factors and their specific receptors in the host. This review will focus on the role of Zn in the crosstalk between the host and microbe, especially for changes in microbial pathogenesis and nociceptive neuron-immune interactions, as it may lead to new ways to prevent or treat microbial infections.


Subject(s)
Host Microbial Interactions/physiology , Host-Pathogen Interactions/physiology , Nociceptors , Zinc/metabolism , Animals , Nociceptors/immunology , Nociceptors/microbiology
19.
AMB Express ; 11(1): 44, 2021 Mar 18.
Article in English | MEDLINE | ID: mdl-33738650

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) F4ac is a major constraint to the development of the pig industry, which is causing newborn and post-weaning piglets diarrhea. Previous studies proved that FaeG is the major fimbrial subunit of F4ac E. coli and efficient for bacterial adherence and receptor recognition. Here we show that the faeG deletion attenuates both the clinical symptoms of F4ac infection and the F4ac-induced intestinal mucosal damage in piglets. Antibody microarray analysis and the detection of mRNA expression using porcine neonatal jejunal IPEC-J2 cells also determined that the absence of FaeG subunit alleviated the F4ac promoted apoptosis in the intestinal epithelial cells. Thus, targeted depletion of FaeG is still beneficial for the prevention or treatment of F4ac infection.

20.
Appl Microbiol Biotechnol ; 104(22): 9719-9732, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33009938

ABSTRACT

Salmonella spp. can cause animal and human salmonellosis. In this study, we established a simple method to detect all Salmonella species by amplifying a specific region within the flgE gene encoding the flagellar hook protein. Our preliminary sequence analysis among flagella-associated genes of Salmonella revealed that although Salmonella Gallinarum and Salmonella Pullorum are lacking flagella, they did have flagella-associated genes, including flgE. To investigate in detail, a comparative flgE sequence analysis was conducted using different bacterial strains including flagellated and non-flagellated Salmonella as well as non-Salmonella strains. Two unique regions (481-529 bp and 721-775 bp of the reference sequence) within the flgE open reading frame were found to be highly conserved and specific to all Salmonella species. Next, we designed a pair of PCR primers (flgE-UP and flgE-LO) targeting the above two regions, and performed a flgE-tailored PCR using as template DNA prepared from a total of 76 bacterial strains (31 flagellated Salmonella strains, 26 non-flagellated Salmonella strains, and 19 other non-Salmonella bacteria strains). Results showed that specific positive bands with expected size were obtained from all Salmonella (including flagellated and non-flagellated Salmonella) strains, while no specific product was generated from non-Salmonella bacterial strains. PCR products from the positive bands were confirmed by DNA sequencing. The minimum detection amount for genomic DNA and bacteria cells reached 18.3 pg/µL and 100 colony-forming unit (CFU) per PCR reaction, respectively. Using the flgE-PCR method to detect Salmonella in artificially contaminated milk samples, as low as 1 CFU/mL Salmonella was detectable after an 8-h pre-culture. Meanwhile, the flgE-tailored PCR method was applied to evaluate 247 clinical samples infected with Salmonella from different chicken breeding farms. The detection results indicated that flgE-PCR could be used to specifically detect Salmonella in concordance with the traditional bacterial culture-based detection method. It is worthwhile noticed that identification results using flgE-tailored PCR should be completed within less than 1 day, expanding the result of much faster than the standard method, which took more than 5 days. Overall, the flgE-tailored PCR method can specifically detect flagellated and non-flagellated Salmonella and can serve as a powerful tool for rapid, simple, and sensitive detection of Salmonella species. KEY POINTS : • Targeting flgE gene for all Salmonella spp. found. • The established PCR assay is used to specifically detect all Salmonella spp. • The PCR method is applied to detect clinical Salmonella spp. samples within less than 1 day.


Subject(s)
Bacterial Proteins , Salmonella , Animals , Bacterial Proteins/genetics , Chickens , Flagella/genetics , Humans , Polymerase Chain Reaction , Salmonella/genetics , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...