Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(2)2024 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-38256103

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales , Proteína S100A12 , Porcinos , Animales , Hibridomas , Calgranulina A , Calgranulina B , Tecnología
2.
Immunology ; 171(3): 365-376, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38013255

RESUMEN

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.


Asunto(s)
Inflamación , Proteína S100A12 , Animales , Humanos , Calgranulina B , Calgranulina A/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo
3.
Int J Mol Sci ; 25(1)2023 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-38203452

RESUMEN

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.


Asunto(s)
Bacterias , Sistemas de Secreción Tipo V , Secreciones Corporales , Agregación Celular , Membrana Celular
4.
Int J Mol Sci ; 23(20)2022 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-36292986

RESUMEN

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.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Zinc , Animales , Zinc/metabolismo , Fosfatidilinositol 3-Quinasas , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Uniones Estrechas , Quinasas MAP Reguladas por Señal Extracelular
5.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216425

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Coinfección/inmunología , Inmunidad Mucosa/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Virosis/inmunología , Animales , Coinfección/microbiología , Coinfección/virología , Humanos , Mucosa Intestinal/virología
6.
Arch Microbiol ; 203(7): 4221-4231, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34091701

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Proteínas de Escherichia coli/metabolismo , Animales , Proteínas de Transporte de Anión/genética , Adhesión Bacteriana/genética , Biopelículas , Línea Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Infecciones por Escherichia coli , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Ratones , Mutación , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Virulencia/genética
7.
J Vis Exp ; (171)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34096912

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales , Antígenos CD13 , Mucosa Intestinal , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Células CHO , Cricetinae , Cricetulus , Epitelio , Escherichia coli , Mucosa Intestinal/inmunología , Porcinos
8.
Appl Microbiol Biotechnol ; 105(13): 5341-5355, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34180006

RESUMEN

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.


Asunto(s)
Antiinfecciosos , Inmunidad Innata , Inmunidad Adaptativa , Animales , Transducción de Señal , Receptores Toll-Like
9.
Vet Res ; 52(1): 39, 2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33663613

RESUMEN

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.


Asunto(s)
Interacciones Microbiota-Huesped/fisiología , Interacciones Huésped-Patógeno/fisiología , Nociceptores , Zinc/metabolismo , Animales , Nociceptores/inmunología , Nociceptores/microbiología
10.
AMB Express ; 11(1): 44, 2021 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-33738650

RESUMEN

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.

11.
Vet Res ; 51(1): 127, 2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028391

RESUMEN

Zinc is the second trace element of living organisms after iron. Given its crucial importance, mammalian hosts restrict the bioavailability of Zinc ions (Zn2+) to bacterial pathogens. As a countermeasure, pathogens utilize high affinity Zn2+ transporters, such as ZnuACB to compete with the host for zinc. It is essential for bacteria to maintain zinc homeostasis and thus maintain their physiology and pathogenesis. In an attempt to uncover the zinc transporter in F4+ enterotoxigenic E. coli (ETEC) C83902, we analyzed two RNA-seq data sets of bacteria samples when different zinc treatments (restriction or abundance) were applied. Considering data revealing that the high affinity zinc uptake system ZnuACB acts as the main transporter in ETEC C83902 to resist zinc deficiency, we deleted znuACB genes to study the role of them in ETEC C83902. The deletion of znuACB genes results in growth perturbation and a sharp decrease in the ability of biofilm formation and adhesion of bacteria in vitro. Taking the data together, this study demonstrates that the ZnuACB system is required for ETEC C83902 to acquire zinc, which highly contributes to ETEC pathogenicity as well.


Asunto(s)
Escherichia coli Enterotoxigénica/fisiología , Infecciones por Escherichia coli/microbiología , Fenotipo , Zinc/metabolismo , Escherichia coli Enterotoxigénica/genética
12.
ACS Appl Bio Mater ; 3(8): 5395-5406, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35021713

RESUMEN

Bacteria's antibiotic resistance is one of the major challenges in the treatment of infectious diseases. With increasing difficulty in discovering antibiotics, there is an urgent need to develop antibiotic-free therapeutic strategies to address this grand challenge. In this report, we developed a polypyrrole (PPy)-based photothermal nano-antibiotic (PTNA) for effective treatment of multidrug-resistant (MDR) bacterial infection. PTNA was fabricated by polymerizing pyrrole onto an anionic vesicle to integrate the cationic and photothermal properties of PPy for combinatory killing against bacteria. PTNA exhibits a strong photothermal effect in the NIR-II (1064 nm) biowindow, thus it is feasible for in vivo therapy due to deeper tissue penetration. Our in vitro experiments revealed that PTNA can significantly inhibit the growth of MDR bacteria (Salmonella typhimurium), alleviate inflammatory response of infected cells, and prevent biofilm formation. More importantly, PTNA showed a significant therapeutic effect in an S. typhimurium-infected animal disease model of acute systemic infection by inhibiting bacterial growth, lowering inflammatory responses and pathological changes, and eventually improving the survival rate of mice. Finally, PTNA had safe profiles in vitro and in vivo with no visible toxicity detected. Therefore, we believe PTNA may serve as a promising antibiotic-free antimicrobial material for the effective treatment of MDR bacterial infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...