Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 358
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 46(1): 38-50, 2017 01 17.
Article in English | MEDLINE | ID: mdl-27986454

ABSTRACT

Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, binds Toll-like receptor 4 (TLR4)-MD2 complex and activates innate immune responses. LPS transfer to TLR4-MD2 is catalyzed by both LPS binding protein (LBP) and CD14. To define the sequential molecular interactions underlying this transfer, we reconstituted in vitro the entire LPS transfer process from LPS micelles to TLR4-MD2. Using electron microscopy and single-molecule approaches, we characterized the dynamic intermediate complexes for LPS transfer: LBP-LPS micelles, CD14-LBP-LPS micelle, and CD14-LPS-TLR4-MD2 complex. A single LBP molecule bound longitudinally to LPS micelles catalyzed multi-rounds of LPS transfer to CD14s that rapidly dissociated from LPB-LPS complex upon LPS transfer via electrostatic interactions. Subsequently, the single LPS molecule bound to CD14 was transferred to TLR4-MD2 in a TLR4-dependent manner. The definition of the structural determinants of the LPS transfer cascade to TLR4 may enable the development of targeted therapeutics for intervention in LPS-induced sepsis.


Subject(s)
Acute-Phase Proteins/immunology , Carrier Proteins/immunology , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 4/immunology , Animals , Humans , Mice , Microscopy, Electron, Transmission , Signal Transduction/immunology
2.
J Proteome Res ; 23(8): 3515-3523, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39007742

ABSTRACT

Acute phase protein (APP) response to vaccine challenges is an attractive alternative to natural infection for identifying pigs with increased disease resilience and monitoring the productive performance. Currently, the methods used for APP quantification are diverse and often based on techniques that use antibodies that are not necessarily pig specific. The objective of this work is the development of a method based on a UPLC-SRM/MS system for simultaneous determination of haptoglobin, apolipoprotein A1, C-reactive protein, pig-major acute protein, and serum amyloid A and its application in pigs to monitor the effect of a vaccine administered against porcine reproductive and respiratory syndrome virus (PRRSV). With the aim of tracing the complete analytical process for each proteotypic peptide, a synthetic QconCat polypeptide construct was designed. It was possible to develop an SRM method including haptoglobin, apolipoprotein A1, pig-MAP, and serum amyloid A1. The PRRSV vaccine only affected haptoglobin. The pigs with positive viremia tended to show higher values than negative pigs, reaching significant differences in the three haptoglobin SRM-detected peptides but not with the data acquired by immunoenzymatic and spectrophotometric assays. These results open the door to the use of SRM to accurately monitor APP changes in experimental pigs.


Subject(s)
Acute-Phase Proteins , Haptoglobins , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Serum Amyloid A Protein , Viral Vaccines , Animals , Swine , Porcine respiratory and reproductive syndrome virus/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine Reproductive and Respiratory Syndrome/immunology , Acute-Phase Proteins/analysis , Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Haptoglobins/analysis , Viral Vaccines/immunology , Serum Amyloid A Protein/analysis , Apolipoprotein A-I/immunology , Apolipoprotein A-I/analysis , C-Reactive Protein/analysis , C-Reactive Protein/immunology , Vaccination , Mass Spectrometry/methods , Viremia/prevention & control , Viremia/immunology
3.
Immunity ; 43(3): 475-87, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26320658

ABSTRACT

Interleukin-17 (IL-17) induces pathology in autoimmunity and infections; therefore, constraint of this pathway is an essential component of its regulation. We demonstrate that the signaling intermediate MCPIP1 (also termed Regnase-1, encoded by Zc3h12a) is a feedback inhibitor of IL-17 receptor signal transduction. MCPIP1 knockdown enhanced IL-17-mediated signaling, requiring MCPIP1's endoribonuclease but not deubiquitinase domain. MCPIP1 haploinsufficient mice showed enhanced resistance to disseminated Candida albicans infection, which was reversed in an Il17ra(-/-) background. Conversely, IL-17-dependent pathology in Zc3h12a(+/-) mice was exacerbated in both EAE and pulmonary inflammation. MCPIP1 degraded Il6 mRNA directly but only modestly downregulated the IL-6 promoter. However, MCPIP1 strongly inhibited the Lcn2 promoter by regulating the mRNA stability of Nfkbiz, encoding the IκBζ transcription factor. Unexpectedly, MCPIP1 degraded Il17ra and Il17rc mRNA, independently of the 3' UTR. The cumulative impact of MCPIP1 on IL-6, IκBζ, and possibly IL-17R subunits results in a biologically relevant inhibition of IL-17 signaling.


Subject(s)
Inflammation/immunology , Interleukin-17/immunology , Ribonucleases/immunology , Signal Transduction/immunology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Candida albicans/immunology , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/immunology , Candidiasis/microbiology , Cell Line , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/immunology , Lipocalins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Oncogene Proteins/metabolism , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Receptors, Interleukin-17/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/genetics , Ribonucleases/metabolism
4.
Immunity ; 39(4): 647-60, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24120359

ABSTRACT

Lipopolysaccharide (LPS) binding protein (LBP) is an acute-phase protein that initiates an immune response after recognition of bacterial LPS. Here, we report the crystal structure of murine LBP at 2.9 Å resolution. Several structural differences were observed between LBP and the related bactericidal/permeability-increasing protein (BPI), and the LBP C-terminal domain contained a negatively charged groove and a hydrophobic "phenylalanine core." A frequent human LBP SNP (allelic frequency 0.08) affected this region, potentially generating a proteinase cleavage site. The mutant protein had a reduced binding capacity for LPS and lipopeptides. SNP carriers displayed a reduced cytokine response after in vivo LPS exposure and lower cytokine concentrations in pneumonia. In a retrospective trial, the LBP SNP was associated with increased mortality rates during sepsis and pneumonia. Thus, the structural integrity of LBP may be crucial for fighting infections efficiently, and future patient stratification might help to develop better therapeutic strategies.


Subject(s)
Acute-Phase Proteins/chemistry , Antimicrobial Cationic Peptides/chemistry , Blood Proteins/chemistry , Carrier Proteins/chemistry , Immunity, Innate/genetics , Lipopolysaccharides/chemistry , Membrane Glycoproteins/chemistry , Models, Molecular , Mutation , Polymorphism, Single Nucleotide , Acute-Phase Proteins/genetics , Acute-Phase Proteins/immunology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Binding Sites , Blood Proteins/genetics , Blood Proteins/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Crystallography, X-Ray , Genotype , Humans , Hydrophobic and Hydrophilic Interactions , Lipopolysaccharides/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Protein Binding , Protein Structure, Tertiary , Static Electricity , Structural Homology, Protein
5.
J Biol Chem ; 295(9): 2570-2581, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31819008

ABSTRACT

Serum amyloid A (SAA), one of the major highly conserved acute-phase proteins in most mammals, is predominantly produced by hepatocytes and also by a variety of cells in extrahepatic tissues. It is well-known that the expression of SAA is sharply increased in bacterial infections. However, the exact physiological function of SAA during bacterial infection remains unclear. Herein, we showed that SAA expression significantly increased in abscesses of Staphylococcus aureus cutaneous infected mice, which exert direct antibacterial effects by binding to the bacterial cell surface and disrupting the cell membrane in acidic conditions. Mechanically, SAA disrupts anionic liposomes by spontaneously forming small vesicles or micelles under acidic conditions. Especially, the N-terminal region of SAA is necessary for membrane disruption and bactericidal activity. Furthermore, we found that mice deficient in SAA1/2 were more susceptible to infection by S. aureus In addition, the expression of SAA in infected skin was regulated by interleukin-6. Taken together, these findings support a key role of the SAA in host defense and may provide a novel therapeutic strategy for cutaneous bacterial infection.


Subject(s)
Anti-Bacterial Agents/metabolism , Immunity, Innate , Serum Amyloid A Protein/metabolism , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/immunology , Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion , Cell Membrane/drug effects , Hydrogen-Ion Concentration , Interleukin-6/physiology , Mice , Serum Amyloid A Protein/immunology , Serum Amyloid A Protein/pharmacology , Staphylococcus aureus/cytology , Staphylococcus aureus/ultrastructure
9.
Rev Med Virol ; 30(5): e2140, 2020 09.
Article in English | MEDLINE | ID: mdl-32686248

ABSTRACT

A knowledge-based cybernetic framework model representing the dynamics of SARS-CoV-2 inside the human body has been studied analytically and in silico to explore the pathophysiologic regulations. The following modeling methodology was developed as a platform to introduce a predictive tool supporting a therapeutic approach to Covid-19 disease. A time-dependent nonlinear system of ordinary differential equations model was constructed involving type-I cells, type-II cells, SARS-CoV-2 virus, inflammatory mediators, interleukins along with host pulmonary gas exchange rate, thermostat control, and mean pressure difference. This formalism introduced about 17 unknown parameters. Estimating these unknown parameters requires a mathematical association with the in vivo sparse data and the dynamic sensitivities of the model. The cybernetic model can simulate a dynamic response to the reduced pulmonary alveolar gas exchange rate, thermostat control, and mean pressure difference under a very critical condition based on equilibrium (steady state) values of the inflammatory mediators and system parameters. In silico analysis of the current cybernetical approach with system dynamical modeling can provide an intellectual framework to help experimentalists identify more active therapeutic approaches.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/immunology , Host-Pathogen Interactions/immunology , Lung/immunology , Nonlinear Dynamics , Pneumonia, Viral/immunology , Acute-Phase Proteins/antagonists & inhibitors , Acute-Phase Proteins/genetics , Acute-Phase Proteins/immunology , Angiotensin-Converting Enzyme 2 , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Betacoronavirus/growth & development , Body Temperature , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/pathology , Coronavirus Infections/virology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Lung/drug effects , Lung/virology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Pulmonary Gas Exchange/drug effects , Pulmonary Gas Exchange/immunology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
10.
Inflammopharmacology ; 29(3): 897-901, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34019218

ABSTRACT

There is an array of plasma protein alterations that occur in a wide variety of species, including humans in response to trauma, inflammation and infections, seemingly irrespective of etiologic agent. In numerous species, these plasma proteins are part of the innate immune response. In addition, it appears that a number of the plasma proteins in this array can be predictive of morbidity and/or mortality. We propose that based on historic use, selected acute phase proteins should be included in ongoing and future non-clinical and clinical studies to help us better understand disease progression in chronic, as well as acute diseases. In addition to assess if there is a relationship between vaccine-induced inflammation and degree of protection from live, attenuated or synthetic vaccines.


Subject(s)
Acute-Phase Proteins/immunology , Infections/immunology , Inflammation/immunology , Wounds and Injuries/immunology , Acute Disease , Animals , Chronic Disease , Humans , Immunity, Innate/immunology
11.
Protein Expr Purif ; 157: 17-20, 2019 05.
Article in English | MEDLINE | ID: mdl-30690139

ABSTRACT

Human lipopolysaccharide (LPS) binding protein (LBP) is a ∼60 kDa glycosylated protein that mediates potent innate immune against invading Gram-negative bacteria by recognition of LPS in their outer membranes. To date, there is no method for efficient production of bioactive LPS-free LBP at sufficient amounts through prokaryotic expression system. Here we present a simple approach for rapid preparation of human LBP from a LPS-eliminated E. coli strain named ClearColi BL21 (DE)3. Combined with the usage of an ultra-high-affinity CL7/Im7 purification system, we achieved one-step purification of recombinant human LBP with over 90% purity at a yield of ∼4 mg/L when using LB culture medium. The produced LBP retains full LPS binding activity which was validated by fluorescence spectroscopy and isothermal titration calorimetry (ITC). Collectively, we develop a valid method that can be applied to cost-effectively produce and purify LPS-free proteins.


Subject(s)
Acute-Phase Proteins/genetics , Carrier Proteins/genetics , Escherichia coli/genetics , Lipopolysaccharides/immunology , Membrane Glycoproteins/genetics , Acute-Phase Proteins/immunology , Acute-Phase Proteins/isolation & purification , Carrier Proteins/immunology , Carrier Proteins/isolation & purification , Cloning, Molecular/methods , Genetic Vectors/genetics , Humans , Lipopolysaccharides/isolation & purification , Membrane Glycoproteins/immunology , Membrane Glycoproteins/isolation & purification , Plasmids/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology
12.
Scand J Rheumatol ; 48(4): 315-319, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30786810

ABSTRACT

Background: Chronic inflammation, as determined by persistently elevated acute-phase reactants in attack-free periods, can occasionally be observed in patients with familial Mediterranean fever (FMF) and is suggested to be a risk factor for the development of amyloidosis. We aimed to investigate the underlying causes of chronic inflammation in FMF patients and its association with amyloidosis in long-term follow-up. Method: Electronic medical records of FMF patients who had regular follow-up for ≥ 5 years in our cohort were utilized. As part of routine evaluation, detailed history, physical examination, and pertinent laboratory and radiographic investigations were performed in all patients to determine potential causes of elevated C-reactive protein (CRP) levels. Results: The study included 146 FMF patients who had no evidence of amyloidosis at baseline and had regular follow-up for ≥ 5 years. Thirty-seven patients (25.3%) were found to have chronic inflammation in the disease course. Twenty-five (67.5%) of them had either very frequent attacks or chronic manifestations of disease. In the entire study group, amyloidosis developed in five patients (3.42%) during the 5 year follow-up, four in the FMF with chronic inflammation group (10.8%), and only one of the 109 patients without chronic inflammation (odds ratio 13.09, 95% confidence interval 1.41-121.2). Conclusions: The results suggest that persistently high CRP levels during the attack-free periods may be a strong risk factor for the development of amyloidosis in patients with FMF. The vast majority of FMF patients with chronic inflammation had active FMF.


Subject(s)
Acute-Phase Proteins/immunology , Amyloidosis , Familial Mediterranean Fever , Inflammation/blood , Adult , Amyloidosis/diagnosis , Amyloidosis/etiology , Amyloidosis/immunology , C-Reactive Protein/analysis , Electronic Health Records/statistics & numerical data , Familial Mediterranean Fever/complications , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/immunology , Female , Follow-Up Studies , Humans , Male , Monitoring, Physiologic/methods , Monitoring, Physiologic/statistics & numerical data , Risk Assessment , Risk Factors
13.
Fish Shellfish Immunol ; 87: 499-506, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30731212

ABSTRACT

Bactericidal permeability-increasing protein (BPI) and lipopolysaccharide-binding protein (LBP) play important roles in host antimicrobial defense. In the present study, we identified one isoform of BPI/LBP gene from turbot (Scophthalmus maximus), designated as SmBPI/LBP1. The full-length cDNA sequence of SmBPI/LBP1 was 1826 bp, which encoding one secreted protein with 480 amino acid residues. Structurally, the SmBPI/LBP1 showed high similarity to its homologs from other vertebrates or invertebrates, which all contained a signal peptide, a BPI/LBP/CETP N-terminal with a LPS-binding domain, and a BPI/LBP/CETP C-terminal domain. The deduced amino acid sequences of SmBPI/LBP1 shared significant similarity to BPI/LBP of Seriola lalandi dorsalis (71%) and Paralichthys olivaceus (69%). Phylogentic analysis further supported that SmBPI/LBP1 act as a new member of vertebrate BPI/LBP family. SmBPI/LBP1 was ubiquitously expressed in all tested tissues, with the highest expression level in spleen tissue. The mRNA expression of SmBPI/LBP1 in spleen and kidney were significantly up-regulated after Vibrio vulnificus challenge. Finally, the recombinant SmBPI/LBP1 showed high affinity to lipopolysaccharide, followed by peptidoglycan and lipoteichoic acid, which is the ubiquitous component of Gram-negative or Gram-positive bacteria. These results indicated that SmBPI/LBP1 probably played important roles in immune response against bacteria infection.


Subject(s)
Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Flatfishes/genetics , Flatfishes/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Acute-Phase Proteins/chemistry , Acute-Phase Proteins/genetics , Acute-Phase Proteins/immunology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Base Sequence , Blood Proteins/chemistry , Blood Proteins/genetics , Blood Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/immunology , Fish Proteins/chemistry , Gene Expression Profiling/veterinary , Lipopolysaccharides/physiology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Peptidoglycan , Phylogeny , Sequence Alignment/veterinary , Teichoic Acids , Vibrio Infections/immunology , Vibrio Infections/veterinary , Vibrio vulnificus/physiology
14.
PLoS Pathog ; 12(12): e1005973, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27977798

ABSTRACT

The host inflammatory response against infections is characterized by the release of pro-inflammatory cytokines and acute-phase proteins, driving both innate and adaptive arms of the immune response. Distinct patterns of circulating cytokines and acute-phase responses have proven indispensable for guiding the diagnosis and management of infectious diseases. This review discusses the profiles of acute-phase proteins and circulating cytokines encountered in viral and bacterial infections. We also propose a model in which the inflammatory response to viral (IL-18/ferritin) and bacterial (IL-6/CRP) infections presents with specific plasma patterns of immune biomarkers.


Subject(s)
Acute-Phase Proteins/immunology , Bacterial Infections/immunology , Cytokines/immunology , Inflammation/immunology , Virus Diseases/immunology , Animals , C-Reactive Protein/immunology , Ferritins/immunology , Humans , Inflammation Mediators/immunology , Interleukin-18/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology
15.
Cytokine ; 103: 90-98, 2018 03.
Article in English | MEDLINE | ID: mdl-28982582

ABSTRACT

Emerging evidence shows that chronic low-grade inflammation and changes in markers of innate immunity are implicated in a range of metabolic abnormalities following an episode of acute pancreatitis. Also, deranged iron metabolism has been linked to type 2 diabetes mellitus, gestational diabetes, and new-onset diabetes after pancreatitis - the conditions characterized by high haemoglobin glycation index (HGI). This study aimed to investigate the associations between markers of innate immunity and iron metabolism in individuals after acute pancreatitis. Fasting blood samples were collected to analyse lipopolysaccharide binding protein (LBP), interleukin (IL)-6, tumor necrosis factor-α, hepcidin, ferritin, soluble transferrin receptor, HbA1c, and glucose. Participants were categorized into two groups: low HGI and high HGI. Linear regression analyses were conducted, and potential confounders (age, sex, ethnicity, body mass index, diabetes mellitus status, smoking status, aetiology of pancreatitis, duration, recurrence, and severity of pancreatitis) were adjusted for in 5 statistical models. A total of 93 patients following an episode of acute pancreatitis were included, of who 40 (43%) had high HGI. In the overall cohort, LBP was significantly associated with hepcidin and ferritin, and IL-6 was significantly associated with hepcidin, consistently in all the models. Further, LBP contributed to 7.7% and 9.5% of variance in hepcidin and ferritin levels, respectively, whereas IL-6 contributed to 5.3% of hepcidin variance. Upon subgroup analysis, the observed LBP associations were maintained in the high HGI subgroup only and the IL-6 association in the low HGI subgroup only. No consistently significant associations were found between any of the other markers. The interplay between LBP, IL-6, hepcidin, and ferritin characterizes metabolic derangements after acute pancreatitis and may play a role in the pathogenesis of new-onset diabetes after pancreatitis.


Subject(s)
Carrier Proteins/blood , Ferritins/immunology , Immunity, Innate , Interleukin-6/blood , Iron/blood , Membrane Glycoproteins/blood , Pancreatitis/blood , Acute Disease , Acute-Phase Proteins/immunology , Adult , Aged , Carrier Proteins/immunology , Cross-Sectional Studies , Female , Ferritins/blood , Hepcidins/immunology , Humans , Interleukin-6/immunology , Iron/immunology , Male , Membrane Glycoproteins/immunology , Middle Aged , Pancreatitis/immunology , Pancreatitis/pathology
16.
J Immunol ; 197(6): 2390-9, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27534554

ABSTRACT

During Gram-negative bacterial infections, excessive LPS induces inflammation and sepsis via action on immune cells. However, the bulk of LPS can be cleared from circulation by the liver. Liver clearance is thought to be a slow process mediated exclusively by phagocytic resident macrophages, Kupffer cells (KC). However, we discovered that LPS disappears rapidly from the circulation, with a half-life of 2-4 min in mice, and liver eliminates about three quarters of LPS from blood circulation. Using microscopic techniques, we found that ∼75% of fluor-tagged LPS in liver became associated with liver sinusoidal endothelial cells (LSEC) and only ∼25% with KC. Notably, the ratio of LSEC-KC-associated LPS remained unchanged 45 min after infusion, indicating that LSEC independently processes the LPS. Most interestingly, results of kinetic analysis of LPS bioactivity, using modified limulus amebocyte lysate assay, suggest that recombinant factor C, an LPS binding protein, competitively inhibits high-density lipoprotein (HDL)-mediated LPS association with LSEC early in the process. Supporting the previous notion, 3 min postinfusion, 75% of infused fluorescently tagged LPS-HDL complex associates with LSEC, suggesting that HDL facilitates LPS clearance. These results lead us to propose a new paradigm of LSEC and HDL in clearing LPS with a potential to avoid inflammation during sepsis.


Subject(s)
Endothelial Cells/physiology , Lipopolysaccharides/blood , Lipopolysaccharides/metabolism , Lipoproteins, HDL/metabolism , Liver/cytology , Acute-Phase Proteins/immunology , Acute-Phase Proteins/metabolism , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Endothelial Cells/immunology , Gram-Negative Bacterial Infections/immunology , Half-Life , Inflammation/immunology , Inflammation/prevention & control , Kinetics , Kupffer Cells/immunology , Lipopolysaccharides/immunology , Lipoproteins, HDL/immunology , Liver/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Sepsis/immunology
17.
Mar Drugs ; 16(5)2018 May 17.
Article in English | MEDLINE | ID: mdl-29772753

ABSTRACT

Recently, accumulating evidence has suggested that Enteromorpha clathrata polysaccharide (ECP) could contribute to the treatment of diseases. However, as a promising candidate for marine drug development, although ECP has been extensively studied, less consideration has been given to exploring its effect on gut microbiota. In this light, given the critical role of gut microbiota in health and disease, we investigated here the effect of ECP on gut microbiota using 16S rRNA high-throughput sequencing. As revealed by bioinformatic analyses, ECP considerably changed the structure of the gut microbiota and significantly promoted the growth of probiotic bacteria in C57BL/6J mice. However, interestingly, ECP exerted different effects on male and female microbiota. In females, ECP increased the abundances of Bifidobacterium spp. and Akkermansia muciniphila, a next-generation probiotic bacterium, whereas in males, ECP increased the population of Lactobacillus spp. Moreover, by shaping a more balanced structure of the microbiota, ECP remarkably reduced the antigen load from the gut in females. Altogether, our study demonstrates for the first time a prebiotic effect of ECP on gut microbiota and forms the basis for the development of ECP as a novel gut microbiota modulator for health promotion and disease management.


Subject(s)
Aquatic Organisms/metabolism , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Polysaccharides/pharmacology , Ulva/metabolism , Acute-Phase Proteins/immunology , Administration, Oral , Animals , Bifidobacterium/drug effects , Bifidobacterium/isolation & purification , Carrier Proteins/blood , Carrier Proteins/immunology , Computational Biology , Dietary Supplements , Disease Models, Animal , Dysbiosis/blood , Dysbiosis/immunology , Female , Humans , Lactobacillus/drug effects , Lactobacillus/isolation & purification , Male , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Polysaccharides/isolation & purification , Polysaccharides/therapeutic use , Specific Pathogen-Free Organisms , Verrucomicrobia/drug effects , Verrucomicrobia/isolation & purification
18.
An Acad Bras Cienc ; 90(3): 2977-2990, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30304229

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is the most common X-linked muscular disease affecting humans. The Golden Retriever Muscular Dystrophy model (GRMD) is considerthe most suitable for several studies. This assay aims to quantify lymphocyte subpopulations CD4, CD5, and CD8, and standardize, the serum electrophoretic profile, to understand their contribution to the pathologic process in normal Golden Retriever dogs (GR group) and dystrophic´s (GRMD group), through the umbilical cord blood, in dogs aged from 2 to 3 months (GR II and GRMD II), and in dogs over 1 year of age (GR III and GRMD III). No significant differences were observed between the CD8+ lymphocyte subpopulations of the groups studied. The CD4+ and CD5+ lymphocyte subpopulations were significantly higher in the GRMD III group compared to the GR III group. Twenty-two different proteins in the gel were identified. The serum concentrations of the proteins belonging to the GR I and GRMD I groups were significantly lower than those of the other groups. We show that expression of acute phase proteins are worst during the aging of the dogs. We hope to expand knowledge to better understand the GRMD model and the translational data.


Subject(s)
Acute-Phase Proteins/immunology , Blood Proteins/immunology , Disease Models, Animal , Immunophenotyping , Muscular Dystrophy, Duchenne/immunology , Age Factors , Animals , Dog Diseases/immunology , Dogs , Flow Cytometry , Inflammation/immunology
19.
Fish Shellfish Immunol ; 60: 436-446, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27979780

ABSTRACT

Lipopolysaccharide-binding protein and bactericidal permeability-increasing protein (LBP/BPI) play crucial role in modulating cellular signals in response to Gram-negative bacteria infection. In the present study, two isoforms of LBP/BPI genes, designated as HcLBP/BPI1 and HcLBP/BPI2, respectively, were cloned from the mussel Hyriopsis cumingii by RACE approach. The full-length cDNA sequences of HcLBP/BPI1 and HcLBP/BPI2 were 1887 and 2227 bp and encoded two secreted proteins of 501 and 518 amino acid residues, respectively. The deduced amino acid of HcLBP/BPI1 and HcLBP/BPI2 contained several conserved domains, such as signal peptide, two BPI/LBP and one central domain. Phylogentic analysis further supported that HcLBP/BPI1 and HcLBP/BPI2 belonged to new members of invertebrate LBP/BPI family. The mRNA transcripts of HcLBP/BPI1 and HcLBP/BPI2 were ubiquitously expressed in all examined tissues, and the expression level of HcLBP/BPI1 was higher than that of HcLBP/BPI2. The mRNA expression of HcLBP/BPI1 in hepatopancreas and hemocytes was significantly up-regulate after Aeromonas hydrophila and LPS challenge, and HcLBP/BPI2 in hepatopancreas was only up-regulated at 6 and 12 h after LPS challenge and at 12 h after A. hydrophila challenge. In addition, the recombinant HcLBP/BPIs displayed antibacterial activity against Gram-negative bacteria, and the antibacterial index of HcLBP/BPI1 was higher than that of HcLBP/BPI2. These results indicated that HcLBP/BPI1 and HcLBP/BPI2 probably played distinct roles in bacterial mediating immune response in Mollusca.


Subject(s)
Acute-Phase Proteins/genetics , Antimicrobial Cationic Peptides/genetics , Blood Proteins/genetics , Carrier Proteins/genetics , Immunity, Innate/genetics , Membrane Glycoproteins/genetics , Unionidae/genetics , Unionidae/immunology , Acute-Phase Proteins/immunology , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/immunology , Base Sequence , Blood Proteins/immunology , Carrier Proteins/immunology , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/immunology , Phylogeny , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Unionidae/classification , Unionidae/microbiology
20.
Int J Mol Sci ; 19(1)2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29283389

ABSTRACT

Staphylococcus aureus is the most common contagious pathogen associated with bovine subclinical mastitis. Current diagnosis of S. aureus mastitis is based on bacteriological culture of milk samples and somatic cell counts, which lack either sensitivity or specificity. Identification of milk proteins that contribute to host defense and their variable responses to pathogenic stimuli would enable the characterization of putative biomarkers of subclinical mastitis. To accomplish this, milk whey samples from healthy and mastitic dairy cows were analyzed using a label-free quantitative proteomics approach. In total, 90 proteins were identified, of which 25 showed significant differential abundance between healthy and mastitic samples. In silico functional analyses indicated the involvement of the differentially abundant proteins in biological mechanisms and signaling pathways related to host defense including pathogen-recognition, direct antimicrobial function, and the acute-phase response. This proteomics and bioinformatics analysis not only facilitates the identification of putative biomarkers of S. aureus subclinical mastitis but also recapitulates previous findings demonstrating the abundance of host defense proteins in intramammary infection. All mass spectrometry data are available via ProteomeXchange with identifier PXD007516.


Subject(s)
Acute-Phase Proteins/genetics , Antimicrobial Cationic Peptides/genetics , Mastitis, Bovine/genetics , Proteome/genetics , Staphylococcal Infections/genetics , Whey Proteins/genetics , Acute-Phase Proteins/immunology , Acute-Phase Proteins/isolation & purification , Animals , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/isolation & purification , Cattle , Female , Mastitis, Bovine/immunology , Protein Interaction Mapping , Proteome/immunology , Proteome/isolation & purification , Proteomics/methods , Staphylococcal Infections/immunology , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Whey/chemistry , Whey Proteins/immunology , Whey Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL