Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Chinese Critical Care Medicine ; (12): 626-629, 2021.
Article in Chinese | WPRIM | ID: wpr-909373

ABSTRACT

Sepsis is a life-threatening organ dysfunction due to the dysregulation of host responses during infection. Severe systemic inflammatory response syndrome (SIRS) is the primary pathophysiological feature. Despite the classical antibiotic therapies play an important role in sepsis, the emergence of multi-resistant bacteria makes a greater challenge in clinical. Antimicrobial peptides (AMP) which consist of small cationic peptides, can be found in most organisms. As a result of their board-spectrum antibacterial activities and immunoregulatory functions, AMPs may have an excellent effect on the treatment of sepsis. In this review, we will discuss the basic role of AMPs in sepsis treatment and their application prospect and the challenges which need to be resolved in order to provide ideas for clinical application of AMPs.

2.
Immune Network ; : e6-2019.
Article in English | WPRIM | ID: wpr-740208

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a unique subset of cells with different functional characteristics compared to classical dendritic cells. The pDCs are critical for the production of type I IFN in response to microbial and self-nucleic acids. They have an important role for host defense against viral pathogen infections. In addition, pDCs have been well studied as a critical player for breaking tolerance to self-nucleic acids that induce autoimmune disorders such as systemic lupus erythematosus. However, pDCs have an immunoregulatory role in inducing the immune tolerance by generating Tregs and various regulatory mechanisms in mucosal tissues. Here, we summarize the recent studies of pDCs that focused on the functional characteristics of gut pDCs, including interactions with other immune cells in the gut. Furthermore, the dynamic role of gut pDCs will be investigated with respect to disease status including gut infection, inflammatory bowel disease, and cancers.


Subject(s)
Dendritic Cells , Immune Tolerance , Inflammatory Bowel Diseases , Interferon Type I , Lupus Erythematosus, Systemic , Mucous Membrane
3.
Chinese Medical Journal ; (24): 1533-1540, 2018.
Article in English | WPRIM | ID: wpr-688080

ABSTRACT

<p><b>Background</b>The previous study showed that mycophenolic acid (MPA) synergizing with lipopolysaccharide (LPS) promoted interleukin (IL)-1β release, but the mechanism is unclear. This study aimed to investigate the mechanism of MPA synergizing with LPS to induce IL-1β release.</p><p><b>Methods</b>Undiluted human blood cells, THP-1 human myeloid leukemia mononuclear cells (THP-1) cells, or monocytes were stimulated with LPS and treated with or without MPA, and the supernatant IL-1β was detected by enzyme-linked immunosorbent assay. The mRNA levels of IL-1β were detected by real-time quantitative polymerase chain reaction. The intracellular protein levels of nuclear factor kappa B (NF-κB) phospho-p65 (p-p65), precursor interleukin-1β (pro-IL-1β), NOD-like receptor pyrin domain containing-3 (NLRP3), and cysteine aspartic acid-specific protease-1 (caspase-1) p20 in THP-1 cell were measured by Western blot.</p><p><b>Results</b>The MPA alone failed to induce IL-1β, whereas MPA synergized with LPS to increase IL-1β in a dose-dependent manner (685.00 ± 20.00 pg/ml in LPS + 5 μmol/L MPA group, P = 0.035; 742.00 ± 31.58 pg/ml in LPS + 25 μmol/L MPA group, P = 0.017; 1000.00 ± 65.59 pg/ml in LPS + 75 μmol/L MPA group, P = 0.024; versus 408.00 ± 35.50 pg/ml in LPS group). MPA alone has no effect on the IL-1β mRNA expression, LPS induced the expression of IL-1β mRNA 2761 fold, and LPS + MPA increased the IL-1β expression 3018 fold, which had the same effect with LPS group (P = 0.834). MPA did not affect the intracellular NF-κB p-p65 and pro-IL-1β protein levels but activated NLRP3 inflammasome. Ac-YVAD-cmk blocked the activation of caspase-1 and subsequently attenuated IL-1β secretion (181.00 ± 45.24 pg/ml in LPS + MPA + YVAD group vs. 588.00 ± 41.99 pg/ml in LPS + MPA group, P = 0.014).</p><p><b>Conclusions</b>Taken together, MPA synergized with LPS to induce IL-1β release via the activation of caspase-1, rather than the enhanced production of pro-IL-1β. These findings suggested that patients immunosuppressed with mycophenolate mofetil may have overly activated caspase-1 during infection, which might contribute to a more sensitive host defense response to invading germs.</p>


Subject(s)
Animals , Humans , Mice , Caspase 1 , Metabolism , Cells, Cultured , Inflammasomes , Interleukin-1beta , Metabolism , Lipopolysaccharides , Pharmacology , Mice, Inbred NOD , Mycophenolic Acid , Pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein
4.
Braz. arch. biol. technol ; 59: e16160450, 2016. tab
Article in English | LILACS | ID: biblio-951371

ABSTRACT

ABSTRACT Computational investigation of a set of publicly available plant microRNAs revealed 19 barley- and other plants-encoded miRNAs and their near-complement reverse sequences (miRNA*) that have potential to bind all B/CYDV open reading frames (ORFs) except ORF0 and ORF6. These miRNAs/miRNAs*, their binding positions and targets are discussed in the context of biological protection of cereals against B/CYDV, based on antiviral silencing.

5.
Immune Network ; : 67-72, 2014.
Article in English | WPRIM | ID: wpr-121976

ABSTRACT

The herpes virus entry mediator (HVEM) is a member of the tumor necrosis factor receptor superfamily (TNFRSF), and therefore it is also known as TNFRSF14 or CD270 (1,2). In recent years, we have focused on understanding HVEM function in the mucosa of the intestine, particularly on the role of HVEM in colitis pathogenesis, host defense and regulation of the microbiota (2,3,4). HVEM is an unusual TNF receptor because of its high expression levels in the gut epithelium, its capacity to bind ligands that are not members of the TNF super family, including immunoglobulin (Ig) superfamily members BTLA and CD160, and its bi-directional functionality, acting as a signaling receptor or as a ligand for the receptor BTLA. Clinically, Hvem recently was reported as an inflammatory bowel disease (IBD) risk gene as a result of genome wide association studies (5,6). This suggests HVEM could have a regulatory role influencing the regulation of epithelial barrier, host defense and the microbiota. Consistent with this, using mouse models, we have revealed how HVEM is involved in colitis pathogenesis, mucosal host defense and epithelial immunity (3,7). Although further studies are needed, our results provide the fundamental basis for understanding why Hvem is an IBD risk gene, and they confirm that HVEM is a mucosal gatekeeper with multiple regulatory functions in the mucosa.


Subject(s)
Animals , Humans , Mice , Colitis , Epithelium , Genome-Wide Association Study , Immune System , Immunity, Innate , Immunity, Mucosal , Immunoglobulins , Inflammation , Inflammatory Bowel Diseases , Intestines , Ligands , Microbiota , Mucous Membrane , Receptors, Tumor Necrosis Factor , Virus Internalization
6.
Journal of Bacteriology and Virology ; : 215-225, 2014.
Article in English | WPRIM | ID: wpr-70874

ABSTRACT

In response to invading pathogens, the body immune system develops an immediate defense mechanism, i.e., innate immune response, which is detected in almost all living organisms including mammals, plants, insects, etc. Recent studies have identified numerous innate immune receptors that are able to recognize pathogen-associated molecular patterns and transduce the essential intracellular signaling cascades to mount early and successful host defenses against infectious challenge. Among innate immune receptors, we will focus on two important receptors, toll-like receptors (TLRs) and nucleotide binding oligomerization domain (Nod)-like receptors, and their major intracellular signaling pathways that culminate to activate innate immune effectors and inflammatory mediators during pathogen infection. In this review, we address the recent advances of understanding intracellular signaling mechanisms by which TLRs and NLRs activate host immune defense and inflammation. The role and regulatory mechanisms by which a subet of NLRs-associated inflammasome activation induce interleukin-1beta secretion and their relevance with host defense will be also discussed. Both TLR- and NLR-mediated intracellular signaling networks serve crucial roles in mounting resistance to bacterial and viral infection through synthesis of immune mediators and antimicrobial chemicals during infection.


Subject(s)
Immune System , Immunity, Innate , Inflammation , Insecta , Interleukin-1beta , Mammals , Toll-Like Receptors
7.
Journal of Korean Medical Science ; : 1415-1423, 2013.
Article in English | WPRIM | ID: wpr-170536

ABSTRACT

The inflammasome is a multi-protein complex that induces maturation of inflammatory cytokines interleukin (IL)-1beta and IL-18 through activation of caspase-1. Several nucleotide binding oligomerization domain-like receptor family members, including NLRP3, recognize unique microbial and danger components and play a central role in inflammasome activation. The NLRP3 inflammasome is critical for maintenance of homeostasis against pathogenic infections. However, inflammasome activation acts as a double-edged sword for various bacterial infections. When the IL-1 family of cytokines is secreted excessively, they cause tissue damage and extensive inflammatory responses that are potentially hazardous for the host. Emerging evidence has shown that diverse bacterial pathogens or their components negatively regulate inflammasome activation to escape the immune response. In this review, we discuss the current knowledge of the roles and regulation of the NLRP3 inflammasome during bacterial infections. Activation and regulation of the NLRP3 inflammasome should be tightly controlled to prevent virulence and pathology during infections. Understanding the roles and regulatory mechanisms of the NLRP3 inflammasome is essential for developing potential treatment approaches against pathogenic infections.


Subject(s)
Humans , Bacterial Infections/immunology , Carrier Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/immunology , Interleukin-1beta/metabolism , Signal Transduction
8.
Article in English | IMSEAR | ID: sea-136415

ABSTRACT

Human rhinoviruses are not only the main pathogens responsible for the common cold, but are now recognized to have a major impact on asthma pathogenesis. There is evidence that rhinovirus infections play a role in asthma development, asthma exacerbations and, potentially, airway remodeling. Children who experience repeated rhinovirus-induced wheezing episodes in infancy have a significantly increased risk of developing asthma, even when compared to children who experience wheezing induced by respiratory syncytial virus. Rhinovirus is also the dominant virus type associated with acute exacerbations of asthma. The epithelial cell is the principal site of rhinovirus infection in both the upper and lower airways and there is strong evidence that virus-induced alterations of epithelial cell biology play a critical role in regulating clinical outcomes. This includes rhinovirus-induced epithelial generation of a variety of chemokines, cytokines and growth factors that likely play a role in viral modulation of airway inflammation. It has also become clear, however, that epithelial cells play an important role in the innate antiviral response to rhinovirus infection, raising the possibility that the relative induction of epithelial host innate antiviral responses versus proinflammatory responses may be one factor regulating the susceptibility of asthmatic subjects to virus-induced disease exacerbations. Recent evidence has also highlighted that rhinovirus infection induces epithelial production of a number of growth factors and other mediators that could contribute to the development and progression of airway remodeling processes in asthma. The current article reviews our current state of knowledge in these areas.

9.
Allergy, Asthma & Immunology Research ; : 81-88, 2011.
Article in English | WPRIM | ID: wpr-163123

ABSTRACT

Interleukin-33 (IL-33) is the 11th member of IL-1 cytokine family which includes IL-1 and IL-18. Unlike IL-1beta and IL-18, IL-33 is suggested to function as an alarmin that is released upon endothelial or epithelial cell damage and may not enhance acquired immune responses through activation of inflammasome. ST2, a IL-33 receptor component, is preferentially expressed by T-helper type (Th) 2 cells, mast cells, eosinophils and basophils, compared to Th1 cells, Th17 cells and neutrophils. Thus, IL-33 profoundly enhances allergic inflammation through increased expression of proallergic cytokines and chemokines. Indeed, IL-33 and its receptor genes are recognized as the most susceptible genes for asthma by several recent genomewide association studies. It has also recently been shown that IL-33 plays a crucial role in innate eosinophilic airway inflammation rather than acquired immune responses such as IgE production. As such, IL-33 provides a unique therapeutic way for asthma, i.e., ameliorating innate airway inflammation.


Subject(s)
Humans , Asthma , Autoimmunity , Basophils , Chemokines , Chronic Disease , Cytokines , Eosinophils , Epithelial Cells , Hypersensitivity , Immunoglobulin E , Inflammation , Interleukin-1 , Interleukin-18 , Mast Cells , Neutrophils , Th1 Cells , Th17 Cells
10.
Infectio ; 14(1): 55-67, mar. 2010. tab
Article in Spanish | LILACS | ID: lil-560933

ABSTRACT

Los péptidos antimicrobianos son las moléculas efectoras del sistema inmune innato, cuyas familias se encuentran en casi todos los organismos, desde bacterias hasta mamíferos. Son una familia de sustancias polifacéticascon complejos mecanismos deacción relacionados con la interacción con el patógeno a través de su membrana, o afectando blancos internos, como la replicación del ADN y la síntesis de proteínas, e interactuando con el huésped con funciones inmunomoduladoras de la regulación delproceso inflamatorio y de la cicatrización. Aunque la generación de resistencia a los péptidos antimicrobianos es mucho menorsi se compara con la generada por losantibióticos convencionales, existen mecanismos de resistencia ya descritos, como la degradación por proteasas, la liberación de proteínas inhibidoras o los cambios en la conformación de la membrana externa del patógeno. El estudio de estas sustancias hapermitido evidenciar sus usos potenciales en el ámbito clínico para contrarrestar los inconvenientes de la resistencia a los antibióticos; sin embargo, a pesar de los grandesavances logrados en este campo, aún quedan puntos controversiales por dilucidar.


The antimicrobial peptides (AMP) are theeffectors molecules of the innate immunesystem, finding groups of this kind of substances in almost all living organisms from bacteria to mammals. They are a family of versatile substances with complexes action mechanisms in the pathogen they interact with membrane, DNA synthesis and protein synthesis and folding, and also with the hostshowing immunomodulatory functions inwound healing and inflammation process.Even though the generation of resistance to the AMP is lower compare with conventional antibiotics there are resistance mechanism already describe to this kind of substances like degradation by proteases, releasing ofinhibitory substances or conformationalchanges in the external membrane of thepathogen. Actually the study of this group of substances has make them see as potential tools for clinical use helping to coun-teract the problem of antibiotic resistance, but even great progress had been made in this field there still exist some controversial issues for future study.


Subject(s)
Cathelicidins , Cecropins , Antimicrobial Cationic Peptides , alpha-Defensins , Anti-Bacterial Agents
11.
Gut and Liver ; : 43-53, 2010.
Article in English | WPRIM | ID: wpr-152061

ABSTRACT

BACKGROUND/AIMS: Loss of transforming growth factor beta1 (TGF-beta1) exhibits a similar pathology to that seen in a subset of individuals infected with Helicobacter pylori, including propagated gastric inflammation, oxidative stress, and autoimmune features. We thus hypothesized that gastric mucosal TGF-beta1 levels could be used to determine the outcome after H. pylori infection. METHODS: Northern blot for the TGF-beta1 transcript, staining of TGF-beta1 expression, luciferase reporter assay, and enzyme-linked immunosorbent assay for TGF-beta1 levels were performed at different times after H. pylori infection. RESULTS: The TGF-beta1 level was markedly lower in patients with H. pylori-induced gastritis than in patients with a similar degree of gastritis induced by nonsteroidal anti-inflammatory drugs. There was a significant negative correlation between the severity of inflammation and gastric mucosal TGF-beta1 levels. SNU-16 cells showing intact TGF-beta signaling exhibited a marked decrease in TGF-beta1 expression, whereas SNU-638 cells defective in TGF-beta signaling exhibited no such decrease after H. pylori infection. The decreased expressions of TGF-beta1 in SNU-16 cells recovered to normal after 24 hr of H. pylori infection, but lasted very spatial times, suggesting that attenuated expression of TGF-beta1 is a host defense mechanism to avoid attachment of H. pylori. CONCLUSIONS: H. pylori infection was associated with depressed gastric mucosal TGF-beta1 for up to 24 hr, but this apparent strategy for rescuing cells from H. pylori attachment exacerbated the gastric inflammation.


Subject(s)
Humans , Blotting, Northern , Enzyme-Linked Immunosorbent Assay , Gastritis , Helicobacter pylori , Inflammation , Luciferases , Oxidative Stress , Transforming Growth Factor beta , Transforming Growth Factor beta1 , Ulcer
12.
Yonsei Medical Journal ; : 1-11, 2009.
Article in English | WPRIM | ID: wpr-83538

ABSTRACT

Mycobacterium tuberculosis (Mtb) is an intracellular pathogen that infects alveolar macrophages following aerosol transmission. Lung macrophages provide a critical intracellular niche that is required for Mtb to establish infection in the human host. This parasitic relationship is made possible by the capacity of Mtb to block phagosome maturation following entry into the host macrophage, creating an environment that supports bacillary replication. Apoptosis is increasingly understood to play a role in host defense against intracellular pathogens including viruses, fungi, protozoa and bacteria. In the last 15 years an understanding of the role that macrophage apoptosis plays in TB has begun to emerge. Here we review the history and current state of the art of this topic and we offer a model of the macrophage-pathogen interaction that takes into the account the complexities of programmed cell death and the relationship between various death signaling pathways and host defense in TB.


Subject(s)
Animals , Humans , Apoptosis/immunology , Macrophages/cytology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology
13.
Immune Network ; : 46-52, 2009.
Article in English | WPRIM | ID: wpr-49350

ABSTRACT

Although tuberculosis poses a significant health threat to the global population, it is a challenge to develop new and effective therapeutic strategies. Nitric oxide (NO) and inducible NO synthase (iNOS) are important in innate immune responses to various intracellular bacterial infections, including mycobacterial infections. It is generally recognized that reactive nitrogen intermediates play an effective role in host defense mechanisms against tuberculosis. In a murine model of tuberculosis, NO plays a crucial role in antimycobacterial activity; however, it is controversial whether NO is critically involved in host defense against Mycobacterium tuberculosis in humans. Here, we review the roles of NO in host defense against murine and human tuberculosis. We also discuss the specific roles of NO in the central nervous system and lung epithelial cells during mycobacterial infection. A greater understanding of these defense mechanisms in human tuberculosis will aid in the development of new strategies for the treatment of disease.


Subject(s)
Humans , Bacterial Infections , Central Nervous System , Defense Mechanisms , Epithelial Cells , Immunity, Innate , Lung , Macrophages , Mycobacterium tuberculosis , Nitric Oxide , Nitric Oxide Synthase , Nitrogen , Tuberculosis
14.
Yonsei Medical Journal ; : 91-97, 1987.
Article in English | WPRIM | ID: wpr-106504

ABSTRACT

The survival of implanted tumor cells in mice which had been treated with interferon in combination with either adriamycin or vincristine was evaluated. While the majority of tumor cells implanted into normal mice failed to survive (52.1 to 63.5%), most of those implanted into mice which had been pretreated with either adriamycin or vincristine survived. If the mice were secondarily treated with interferon, the ability of adriamycin or vincristine to inhibit the survival of implanted tumor cells was restored within 24 hours. Restoration of tumoricidal activity by interferon treatment was more evident in the adriamycin pretreated mice. Peritoneal macrophages isolated from mice pretreated with both interferon and adriamycin had an increased tumoricidal activity, when compared with those isolated from mice treated with adriamycin alone. This interferon dependent enhancement of tumoricidal activity was comparable with that obtained by treating mice with lymphokines a product of Con A treated lymphocytes isolated from BCG treated mice. These results suggested that both adriamycin and vincristine may damage the macrophages required for the natural host defense mechanism and allow the implanted tumor cells to survive. Interferon may, however, protect the macrophages from drug induced damage.


Subject(s)
Mice , Animals , Doxorubicin/therapeutic use , Interferon Type I/therapeutic use , Macrophages/immunology , Mice, Inbred ICR , Neoplasms, Experimental/therapy , Vincristine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL