Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 12(5): 1643-1667, 2021.
Article in English | MEDLINE | ID: mdl-34242820

ABSTRACT

BACKGROUND & AIMS: Gut microbiota have been reported to be sensitive to circadian rhythms and host lipometabolism, respectively. Although melatonin-mediated beneficial efforts on many physiological sites have been revealed, the regulatory actions of oral melatonin on the communication between gut microbiota and host are still not clear. Angiopoietin-like 4 (ANGPTL4) has been shown to be strongly responsible for the regulation of systemic lipid metabolism. Herein, we identified that oral melatonin improved lipid dysmetabolism in ileum and epididymal white adipose tissue (eWAT) via gut microbiota and ileac ANGPTL4. METHODS: Analyses of jet-lag (JL) mice, JL mice with oral melatonin administration (JL+MT), and the control for mRNA and protein expression regarding lipid uptake and accumulation in ileum and eWAT were made. Gut microbiome sequencing and experimental validation of target strains were included. Functional analysis of key factors/pathways in the various rodent models, including the depletion of gut microbiota, mono-colonization of Escherichia coli, and other genetic intervention was made. Analyses of transcriptional regulation and effects of melatonin on E coli-derived lipopolysaccharide (LPS) in vitro were made. RESULTS: JL mice have a higher level of ileal lipid uptake, fat accumulation in eWAT, and lower level of circulating ANGPTL4 in comparison with the control mice. JL mice also showed a significantly higher abundance of E coli and LPS than the control mice. Conversely, oral melatonin supplementation remarkably reversed these phenotypes. The test of depletion of gut microbiota further demonstrated that oral melatonin-mediated improvements on lipometabolism in JL mice were dependent on the presence of gut microbiota. By mono-colonization of E coli, LPS has been determined to trigger these changes similar to JL. Furthermore, we found that LPS served as a pivotal link that contributed to activating toll-like receptor 4 (TLR4)/signal transducer and activator of transcription 3 (STAT3_/REV-ERBα) signaling to up-regulate nuclear factor interleukin-3-regulated protein (NFIL3) expression, resulting in increased lipid uptake in ileum. In MODE-K cells, the activation of NFIL3 has further been shown to inhibit ANGPTL4 transcription, which is closely associated with lipid uptake and transport in peripheral tissues. Finally, we confirmed that melatonin inhibited LPS via repressing the expression of LpxC in E coli. CONCLUSIONS: Overall, oral melatonin decreased the quantity of E coli-generated LPS, which alleviated NFIL3-induced transcriptional inhibition of ANGPTL4 through TLR4/IL-22/STAT3 signaling in ileum, thereby resulting in the amelioration of ileal lipid intake and lower fat accumulation in eWAT. These results address a novel regulation of oral melatonin originating from gut microbiota to host distal tissues, suggesting that microbe-generated metabolites are potential therapies for melatonin-mediated improvement of circadian rhythm disruption and related metabolic syndrome.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Intestines/drug effects , Intestines/metabolism , Lipid Metabolism/drug effects , Lipopolysaccharides/immunology , Melatonin/administration & dosage , Administration, Oral , Animals , Biomarkers , Circadian Rhythm/drug effects , Escherichia coli/immunology , Gastrointestinal Microbiome/drug effects , Ileum , Mice , Signal Transduction , Toll-Like Receptor 4/metabolism
2.
Front Immunol ; 12: 641696, 2021.
Article in English | MEDLINE | ID: mdl-34079540

ABSTRACT

The impact of antibiotic use for growth promotion in livestock and poultry production on the rise of antimicrobial resistance (AMR) in bacteria led to the ban of this practice in the European Union in 2006 and a restriction of antimicrobial use (AMU) in animal agriculture in Canada and the United States of America. There is a high risk of infectious diseases such as necrotic enteritis due to Clostridium perfringens, and colibacillosis due to avian pathogenic Escherichia coli in antimicrobial-free broiler chickens. Thus, efficient and cost-effective methods for reducing AMU, maintaining good poultry health and reducing public health risks (food safety) are urgently needed for poultry production. Several alternative agents, including plant-derived polyphenolic compounds, have been investigated for their potential to prevent and control diseases through increasing poultry immunity. Many studies in humans reported that plant flavonoids could modulate the immune system by decreasing production of pro-inflammatory cytokines, T-cell activation, and proliferation. Fruits, especially berries, are excellent sources of flavonoids while being rich in nutrients and other functionally important molecules (vitamins and minerals). Thus, fruit byproducts or wastes could be important resources for value-added applications in poultry production. In the context of the circular economy and waste reduction, this review summarizes observed effects of fruit wastes/extracts on the general health and the immunity of poultry.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Chickens/immunology , Escherichia coli Infections/prevention & control , Fruit/chemistry , Plant Extracts/therapeutic use , Poultry Diseases/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Chickens/growth & development , Clostridium perfringens/immunology , Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Plant Extracts/chemistry , Poultry Diseases/immunology , Poultry Diseases/microbiology
3.
mSphere ; 6(2)2021 03 31.
Article in English | MEDLINE | ID: mdl-33789939

ABSTRACT

Human rotavirus (HRV) infection is a major cause of gastroenteritis in children worldwide. Broad-spectrum antibiotic-induced intestinal microbial imbalance and the ensuing immune-metabolic dysregulation contribute to the persistence of HRV diarrhea. Escherichia coli Nissle 1917 (EcN), a Gram-negative probiotic, was shown to be a potent immunostimulant and alleviated HRV-induced diarrhea in monocolonized gnotobiotic (Gn) piglets. Our goal was to determine how EcN modulates immune responses in ciprofloxacin (Cipro)-treated Gn piglets colonized with a defined commensal microbiota (DM) and challenged with virulent HRV (VirHRV). Cipro given in therapeutic doses for a short term reduced serum and intestinal total and HRV-specific antibody titers, while EcN treatment alleviated this effect. Similarly, EcN treatment increased the numbers of total immunoglobulin-secreting cells, HRV-specific antibody-secreting cells, activated antibody-forming cells, resting/memory antibody-forming B cells, and naive antibody-forming B cells in systemic and/or intestinal tissues. Decreased levels of proinflammatory but increased levels of immunoregulatory cytokines and increased frequencies of Toll-like receptor-expressing cells were evident in the EcN-treated VirHRV-challenged group. Moreover, EcN treatment increased the frequencies of T helper and T cytotoxic cells in systemic and/or intestinal tissues pre-VirHRV challenge and the frequencies of T helper cells, T cytotoxic cells, effector T cells, and T regulatory cells in systemic and/or intestinal tissues postchallenge. Moreover, EcN treatment increased the frequencies of systemic and mucosal conventional and plasmacytoid dendritic cells, respectively, and the frequencies of systemic natural killer cells. Our findings demonstrated that Cipro use altered immune responses of DM-colonized neonatal Gn pigs, while EcN supplementation rescued these immune parameters partially or completely.IMPORTANCE Rotavirus (RV) is a primary cause of malabsorptive diarrhea in children and is associated with significant morbidity and mortality, especially in developing countries. The use of antibiotics exacerbates intestinal microbial imbalance and results in the persistence of RV-induced diarrhea. Probiotics are now being used to treat enteric infections and ulcerative colitis. We showed previously that probiotics partially protected gnotobiotic (Gn) piglets against human RV (HRV) infection and decreased the severity of diarrhea by modulating immune responses. However, the interactions between antibiotic and probiotic treatments and HRV infection in the context of an established gut microbiota are poorly understood. In this study, we developed a Gn pig model to study antibiotic-probiotic-HRV interactions in the context of a defined commensal microbiota (DM) that mimics aspects of the infant gut microbiota. Our results provide valuable information that will contribute to the treatment of antibiotic- and/or HRV-induced diarrhea and may be applicable to other enteric infections in children.


Subject(s)
Adaptive Immunity , Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/therapeutic use , Escherichia coli/immunology , Gastrointestinal Microbiome/drug effects , Immunity, Innate , Probiotics/administration & dosage , Rotavirus Infections/prevention & control , Age Factors , Animals , Cytokines/immunology , Disease Models, Animal , Escherichia coli/classification , Humans , Rotavirus/immunology , Rotavirus Infections/immunology , Swine
4.
Front Immunol ; 11: 1715, 2020.
Article in English | MEDLINE | ID: mdl-32849605

ABSTRACT

Monocytes can develop immunological memory, a functional characteristic widely recognized as innate immune training, to distinguish it from memory in adaptive immune cells. Upon a secondary immune challenge, either homologous or heterologous, trained monocytes/macrophages exhibit a more robust production of pro-inflammatory cytokines, such as IL-1ß, IL-6, and TNF-α, than untrained monocytes. Candida albicans, ß-glucan, and BCG are all inducers of monocyte training and recent metabolic profiling analyses have revealed that training induction is dependent on glycolysis, glutaminolysis, and the cholesterol synthesis pathway, along with fumarate accumulation; interestingly, fumarate itself can induce training. Since fumarate is produced by the tricarboxylic acid (TCA) cycle within mitochondria, we asked whether extra-mitochondrial fumarate has an effect on mitochondrial function. Results showed that the addition of fumarate to monocytes induces mitochondrial Ca2+ uptake, fusion, and increased membrane potential (Δψm), while mitochondrial cristae became closer to each other, suggesting that immediate (from minutes to hours) mitochondrial activation plays a role in the induction phase of innate immune training of monocytes. To establish whether fumarate induces similar mitochondrial changes in vivo in a multicellular organism, effects of fumarate supplementation were tested in the nematode worm Caenorhabditis elegans. This induced mitochondrial fusion in both muscle and intestinal cells and also increased resistance to infection of the pharynx with E. coli. Together, these findings contribute to defining a mitochondrial signature associated with the induction of innate immune training by fumarate treatment, and to the understanding of whole organism infection resistance.


Subject(s)
Caenorhabditis elegans/drug effects , Escherichia coli Infections/prevention & control , Escherichia coli/pathogenicity , Fumarates/pharmacology , Immunity, Innate/drug effects , Immunologic Memory/drug effects , Mitochondria/drug effects , Monocytes/drug effects , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Calcium Signaling/drug effects , Cells, Cultured , Cytokines/metabolism , Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/immunology , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Monocytes/immunology , Monocytes/metabolism
5.
Sci Rep ; 10(1): 12414, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709982

ABSTRACT

The increase of antimicrobial resistance (AMR), and lack of new classes of licensed antimicrobials, have made alternative treatment options for AMR pathogens increasingly attractive. Recent studies have demonstrated anti-bacterial efficacy of a humanised monoclonal antibody (mAb) targeting the O25b O-antigen of Escherichia coli ST131. To evaluate the phenotypic effects of antibody binding to diverse clinical E. coli ST131 O25b bacterial isolates in high-throughput, we designed a novel mAb screening method using high-content imaging (HCI) and image-based morphological profiling to screen a mAb targeting the O25b O-antigen. Screening the antibody against a panel of 86 clinical E. coli ST131 O25:H4 isolates revealed 4 binding phenotypes: no binding (18.60%), weak binding (4.65%), strong binding (69.77%) and strong agglutinating binding (6.98%). Impaired antibody binding could be explained by the presence of insertion sequences or mutations in O-antigen or lipopolysaccharide core biosynthesis genes, affecting the amount, structure or chain length of the O-antigen. The agglutinating binding phenotype was linked with lower O-antigen density, enhanced antibody-mediated phagocytosis and increased serum susceptibly. This study highlights the need to screen candidate mAbs against large panels of clinically relevant isolates, and that HCI can be used to evaluate mAb binding affinity and potential functional efficacy against AMR bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli/drug effects , High-Throughput Screening Assays/methods , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/ultrastructure , Escherichia coli Infections/microbiology , Feasibility Studies , Humans , Interspersed Repetitive Sequences/genetics , Microbial Sensitivity Tests , Microscopy, Electron , O Antigens/genetics , O Antigens/immunology , Phylogeny , Polymorphism, Single Nucleotide , Virulence/immunology
6.
Front Immunol ; 11: 612775, 2020.
Article in English | MEDLINE | ID: mdl-33679699

ABSTRACT

A steady rise in the number of poly-sensitized patients has increased the demand for effective prophylactic strategies against multi-sensitivities. Probiotic bacteria have been successfully used in clinics and experimental models to prevent allergic mono-sensitization. In the present study, we have investigated whether probiotic bacteria could prevent poly-sensitization by imprinting on the immune system early in life. We used two recombinant variants of probiotic Escherichia coli Nissle 1917 (EcN): i) EcN expressing birch and grass pollen, poly-allergen chimera construct (EcN-Chim), and ii) an "empty" EcN without allergen expression (EcN-Ctrl). Conventional mice (CV) were treated with either EcN-Chim or EcN-Ctrl in the last week of the gestation and lactation period. Gnotobiotic mice received one oral dose of either EcN-Chim or EcN-Ctrl before mating. The offspring from both models underwent systemic allergic poly-sensitization and intranasal challenge with recombinant birch and grass pollen allergens (rBet v 1, rPhl p 1, and rPhl p 5). In the CV setting, the colonization of offspring via treatment of mothers reduced allergic airway inflammation (AAI) in offspring compared to poly-sensitized controls. Similarly, in a gnotobiotic model, AAI was reduced in EcN-Chim and EcN-Ctrl mono-colonized offspring. However, allergy prevention was more pronounced in the EcN-Ctrl mono-colonized offspring as compared to EcN-Chim. Mono-colonization with EcN-Ctrl was associated with a shift toward mixed Th1/Treg immune responses, increased expression of TLR2 and TLR4 in the lung, and maintained levels of zonulin-1 in lung epithelial cells as compared to GF poly-sensitized and EcN-Chim mono-colonized mice. This study is the first one to establish the model of allergic poly-sensitization in gnotobiotic mice. Using two different settings, gnotobiotic and conventional mice, we demonstrated that an early life intervention with the EcN without expressing an allergen is a powerful strategy to prevent poly-sensitization later in life.


Subject(s)
Epithelial Cells/immunology , Escherichia coli/immunology , Homeostasis/immunology , Hypersensitivity/immunology , Immune System/immunology , Allergens/immunology , Animals , Antigens, Plant/immunology , Betula/immunology , Female , Germ-Free Life/immunology , Mice , Mice, Inbred BALB C , Poaceae/immunology , Pollen/immunology , Probiotics/administration & dosage
7.
Mol Immunol ; 116: 199-207, 2019 12.
Article in English | MEDLINE | ID: mdl-31731097

ABSTRACT

A 38 kDa ß-1,3-glucanase allergen from Cryptomeria japonica pollen (CJP38) was recombinantly produced in E. coli and purified to homogeneity with the use of Ni-affinity resin. CJP38 hydrolyzed ß-1,3-glucans such as CM-curdlan and laminarioligosaccharides in an endo-splitting manner. The optimum pH and temperature for ß-1,3-glucanase activity were approximately 4.5 and 50 °C, respectively. The enzyme was stable at 30-60 °C and pH 4.0-10.5. Furthermore, CJP38 catalyzed a transglycosylation reaction to yield reaction products with a molecular weight higher than those of the starting laminarioligosaccharide substrates. The three-dimensional structure of CJP38 was determined using X-ray crystallography at 1.5 Å resolution. CJP38 exhibited the typical (ß/α)8 TIM-barrel motif, similar to allergenic ß-1,3-glucanases from banana (Mus a 5) and rubber tree latex (Hev b 2). Amino acid sequence alignment of these proteins indicated that the two-consensus IgE epitopes identified on the molecular surfaces of Mus a 5 and Hev b 2 were highly conserved in CJP38. Their conformations and surface locations were quite similar for these proteins. Sequence and structural conservation of these regions suggest that CJP38 is a candidate allergen responsible for the pollen-latex-fruit syndrome relating to Japanese cedar pollinosis.


Subject(s)
Allergens/chemistry , Antigens, Plant/chemistry , Cryptomeria/chemistry , Pollen/chemistry , Allergens/immunology , Amino Acid Sequence , Antigens, Plant/immunology , Cross Reactions/immunology , Cryptomeria/immunology , Crystallography, X-Ray/methods , Epitopes/chemistry , Epitopes/immunology , Escherichia coli/immunology , Humans , Hydrogen-Ion Concentration , Immunoglobulin E/chemistry , Immunoglobulin E/immunology , Latex/chemistry , Latex/immunology , Musa/chemistry , Musa/immunology , Plant Proteins/chemistry , Plant Proteins/immunology , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Temperature
8.
Mol Immunol ; 114: 19-29, 2019 10.
Article in English | MEDLINE | ID: mdl-31326654

ABSTRACT

Worldwide, more than one-third of the population suffers from allergies. A significant fraction of officially registered allergens originate from the profilin family of proteins. Profilins are small ubiquitous proteins which are found in plants, viruses and various eukaryotes including mammals. Although they are primarily regarded as minor allergens, profilins are important players in immunoglobulin E (IgE) cross-reactivity. However, in some populations profilins are recognized by IgE from at least 50% of patients allergic to a given allergen source. Cuc m 2.0101 is recognized by IgE in more than 80% of muskmelon-allergic patients. The recombinant isoallergen Cuc m 2.0101 was produced in significant quantities and its X-ray crystal structure was determined. In addition, a new Art v 4.0101 (mugwort profilin) structure was determined. The profilins Cuc m 2.0101 and Art v 4.0101 were compared in terms of their structure and thermal stability. Furthermore, structural similarities and IgE cross-reactivity between profilins from different sources are discussed to explain the molecular basis of various clinical syndromes involving this group of allergens. Special emphasis is placed on discussion of profilins' quaternary structures and their relation to biological function, as well as to protein allergenicity. Moreover, a potential impact of protein purification protocols on the structure of profilins is highlighted.


Subject(s)
Antigens, Plant/chemistry , Profilins/chemistry , Amino Acid Sequence , Antigens, Plant/immunology , Cross Reactions/immunology , Escherichia coli/immunology , Escherichia coli/metabolism , Hypersensitivity/immunology , Immunoglobulin E/chemistry , Plant Proteins/chemistry , Plant Proteins/immunology , Pollen/chemistry , Pollen/immunology , Profilins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
9.
J Innate Immun ; 11(2): 150-167, 2019.
Article in English | MEDLINE | ID: mdl-30368505

ABSTRACT

Recent studies demonstrated that allograft inflammatory factor-1 (AIF-1) and RNASET2 act as chemoattractants for macrophages and modulate the inflammatory processes in both vertebrates and invertebrates. The expression of these proteins significantly increases after bacterial infection; however, the mechanisms by which they regulate the innate immune response are still poorly defined. Here, we evaluate the effect of bacterial lipopolysaccharide injection on the expression pattern of these genes and the interrelation between them during innate immune response in the medicinal leech, an invertebrate model with a simple anatomy and a marked similarity with vertebrates in inflammatory processes. Collectively, prokaryotic-eukaryotic co-cultures and in vivo infection assays suggest that RNASET2 and AIF-1 play a crucial role in orchestrating a functional cross-talk between granulocytes and macrophages in leeches, resulting in the activation of an effective response against pathogen infection. RNASET2, firstly released by granulocytes, likely plays an early antibacterial role. Subsequently, AIF-1+ RNASET2-recruited macrophages further recruit other macrophages to potentiate the antibacterial inflammatory response. These experimental data are in keeping with the notion of RNA-SET2 acting as an alarmin-like molecule whose role is to locally transmit a "danger" signal (such as a bacterial infection) to the innate immune system in order to trigger an appropriate host response.


Subject(s)
Calcium-Binding Proteins/metabolism , Endoribonucleases/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Hirudo medicinalis/immunology , Macrophages/immunology , Neutrophils/immunology , Alarmins/metabolism , Animals , Cells, Cultured , Immunity, Innate , Lipopolysaccharides/immunology
10.
Front Immunol ; 10: 3119, 2019.
Article in English | MEDLINE | ID: mdl-32082302

ABSTRACT

Gum arabic (GA) is a traditional herbal medicine from Acacia Senegal (L.) Willdenow trees, which consist of a complex mixture of polysaccharides and glycoproteins. It is used in daily applications for several diseases and is considered to protect against bacterial infections. The detailed mechanisms behind these observations are still unclear. In this study, we investigated the direct antibacterial activity of GA water and ethanol extracts against Staphylococcus (S.) aureus or Escherichia (E.) coli and the immunomodulating properties of those extracts on granulocytes as a first line of defense against bacteria. Firstly, the direct antimicrobial effect of GA was tested on three different S. aureus strains and two E. coli strains. The growth of bacteria was analyzed in the presence of different GA concentrations over time. GA water as well as ethanol extracts showed a significant growth inhibition in a concentration-dependent manner in the case of S. aureus Newman, S. aureus Rd5, and E. coli 25922, but not in the case of S. aureus USA300 and E. coli K1. Transmission electron microscopic analysis confirmed an antibacterial effect of GA on the bacteria. Secondly, the immunomodulatory effect of GA on the antimicrobial activity of bovine or human blood-derived granulocytes was evaluated. Interestingly, water and ethanol extracts enhanced antimicrobial activity of granulocytes by the induction of intracellular ROS production. In line with these data, GA increased the phagocytosis rate of E. coli. No effect was seen on neutrophil extracellular trap (NET) formation that mediates killing of extracellular bacteria such as S. aureus. In conclusion, we show that GA exhibits a direct antibacterial effect against some S. aureus and E. coli strains. Furthermore, GA boosts the antimicrobial activities of granulocytes and increases intracellular ROS production, which may lead to more phagocytosis and intracellular killing. These data might explain the described putative antimicrobial activity of GA used in traditional medicine.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/immunology , Granulocytes/drug effects , Granulocytes/immunology , Gum Arabic/pharmacology , Immunologic Factors/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Animals , Anti-Bacterial Agents/chemistry , Cattle , Dose-Response Relationship, Drug , Escherichia coli/ultrastructure , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Granulocytes/metabolism , Gum Arabic/chemistry , Humans , Immunologic Factors/chemistry , Microbial Sensitivity Tests , Polysaccharides, Bacterial/immunology , Reactive Oxygen Species , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/ultrastructure
11.
J Control Release ; 286: 20-32, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30017722

ABSTRACT

One of the strategies used to improve the immunogenicity of purified protein antigens has relied on their association with synthetic nanocarriers, which, in general, have functioned as simple antigen containers. Here, we present a more advanced strategy based on the design of an antigen nanocarrier at the molecular level. The nanocarrier is composed of a vitamin E oily core, surrounded by two layers: a first layer of chitosan and a second of dextran sulphate. The selected antigen, IutA protein from Escherichia coli, was harboured between the two polymeric layers. The final bilayer nanocapsules had a nanometric size (≈ 200 nm), a negative zeta potential (< -40 mV) and a good antigen association efficiency (≈ 70%). The bilayer architecture led to an improvement on the formulation stability and the controlled release of the associated antigen. Remarkably, after being administered to mice, bilayer nanocapsules elicited higher IgG levels than those obtained with antigen precipitated with Alum. Moreover, freeze-dried nanocapsules were stable at room temperature for, at least, 3 months. These promising data, in addition to their contribution to the development of an uropathogenic E. coli vaccine, has allowed us to validate these novel bilayer nanocapsules as adequate platforms for the delivery of protein antigens.


Subject(s)
Adjuvants, Immunologic/pharmacology , Delayed-Action Preparations/pharmacology , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/administration & dosage , Escherichia coli/immunology , Vitamin E/pharmacology , Adjuvants, Immunologic/chemistry , Animals , Chitosan/chemistry , Chitosan/pharmacology , Delayed-Action Preparations/chemistry , Dextrans/chemistry , Dextrans/pharmacology , Escherichia coli Infections/blood , Escherichia coli Infections/immunology , Escherichia coli Vaccines/pharmacology , Female , Freeze Drying , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Nanocapsules/chemistry , RAW 264.7 Cells , Vitamin E/chemistry
12.
J Surg Res ; 227: 44-51, 2018 07.
Article in English | MEDLINE | ID: mdl-29804861

ABSTRACT

BACKGROUND: Nutritional management is crucial during the acute phase of severe illnesses. However, the appropriate nutritional requirements for patients with sepsis are poorly understood. We investigated alterations in carbohydrate, fat, and protein metabolism in mice with different degrees of sepsis. MATERIALS AND METHODS: C57BL/6 mice were divided into three groups: control mice group, administered with saline, and low- and high-dose lipopolysaccharide (LPS) groups, intraperitoneally administered with 1 and 5 mg of LPS/kg, respectively. Rectal temperature, food intake, body weight, and spontaneous motor activity were measured. Indirect calorimetry was performed using a respiratory gas analysis for 120 h, after which carbohydrate oxidation and fatty acid oxidation were calculated. Urinary nitrogen excretion was measured to evaluate protein metabolism. The substrate utilization ratio was recalculated. Plasma and liver carbohydrate and lipid levels were evaluated at 24, 72, and 120 h after LPS administration. RESULTS: Biological reactions decreased significantly in the low- and high-LPS groups. Fatty acid oxidation and protein oxidation increased significantly 24 h after LPS administration, whereas carbohydrate oxidation decreased significantly. Energy substrate metabolism changed from glucose to predominantly lipid metabolism depending on the degree of sepsis, and protein metabolism was low. Plasma lipid levels decreased, whereas liver lipid levels increased at 24 h, suggesting that lipids were transported to the liver as the energy source. CONCLUSIONS: Our findings revealed that energy substrate metabolism changed depending on the degree of sepsis. Therefore, in nutritional management, such metabolic alterations must be considered, and further studies on the optimum nutritional intervention during severe sepsis are necessary.


Subject(s)
Energy Metabolism , Glucose/metabolism , Lipid Metabolism , Sepsis/metabolism , Animals , Body Weight , Calorimetry, Indirect , Disease Models, Animal , Eating , Escherichia coli/immunology , Humans , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Sepsis/diagnosis , Sepsis/diet therapy , Sepsis/immunology , Severity of Illness Index
13.
Food Funct ; 9(6): 3338-3343, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29808837

ABSTRACT

The objective of this study was to determine the effects of dietary ß-glucan (BG) on growth performance and blood parameters in weaned pigs administered with Escherichia coli lipopolysaccharide (LPS). Twenty four pigs [24 ± 2 days old; 6.60 ± 0.04 kg body weight (BW)] were randomly allocated into two groups (12 pigs per group) with diets supplemented with 0 or BG at 200 mg kg-1 diet (CON vs. BG). These pigs were fed for a 35-day trial. On day 36, six pigs each from CON and BG were intramuscularly administered LPS (50 µg kg-1), while another 6 pigs from CON were intramuscularly administered an equivalent amount of sterile saline. Blood samples were collected at 3 h and rectal temperature data were collected at 0, 4, 8 and 24 h after LPS administration. Results showed that the pigs fed with BG diet had an increased average daily gain in rectal temperature during week 4, week 5 and the overall period, compared with the pigs fed with CON diet (P < 0.05), and resulted in greater final BW (P < 0.05). LPS administration increased the rectal temperature of the pigs fed with CON diet at 4, 8 and 24 h post administration (P < 0.05), and also increased the serum concentrations of pig-major acute phase protein, haptoglobin, tumor necrosis factor-α and interleukine-1 beta (P < 0.05). However, the pigs fed with BG diet had higher concentration of serum complement 3 (P < 0.05) and lower concentration of serum Pig-MAP, HP and interleuking-6 (P = 0.08) compared to that of pigs fed with CON diet after the LPS administration. Moreover, relative to the non-administered pigs, LPS administration increased the concentrations of serum creatinine, direct bilirubin and some of the amino acids in pigs after LPS administration (P < 0.05). In conclusion, the study suggested that feeding BG diet could improve the growth performance and partially alleviate the inflammation response of pigs after LPS administration.


Subject(s)
Dietary Supplements/analysis , Lipopolysaccharides/immunology , Swine/growth & development , Swine/metabolism , beta-Glucans/metabolism , Animal Feed/analysis , Animals , Escherichia coli/chemistry , Escherichia coli/immunology , Female , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Swine/genetics , Swine/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Weaning
14.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563331

ABSTRACT

While the treatment of inflammatory disorders is generally based on inhibiting factors that drive onset of inflammation, these therapies can compromise healing (NSAIDs) or dampen immunity against infections (biologics). In search of new antiinflammatories, efforts have focused on harnessing endogenous pathways that drive resolution of inflammation for therapeutic gain. Identification of specialized pro-resolving mediators (SPMs) (lipoxins, resolvins, protectins, maresins) as effector molecules of resolution has shown promise in this regard. However, their action on inflammatory resolution in humans is unknown. Here, we demonstrate using a model of UV-killed Escherichia coli-triggered skin inflammation that SPMs are biosynthesized at the local site at the start of resolution, coinciding with the expression of receptors that transduce their actions. These include receptors for lipoxin A4 (ALX/FPR2), resolvin E1 (ChemR23), resolvin D2 (GPR18), and resolvin D1 (GPR32) that were differentially expressed on the endothelium and infiltrating leukocytes. Administering SPMs into the inflamed site 4 hours after bacterial injection caused a reduction in PMN numbers over the ensuing 6 hours, the phase of active resolution in this model. These results indicate that in humans, the appearance of SPMs and their receptors is associated with the beginning of inflammatory resolution and that their therapeutic supplementation enhanced the resolution response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Escherichia coli/immunology , Inflammation/immunology , Inflammation/metabolism , Skin/immunology , Skin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Blister/immunology , Blister/metabolism , Chemokines/metabolism , Cytokines/metabolism , Docosahexaenoic Acids/pharmacology , Eicosanoids/immunology , Eicosanoids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Escherichia coli/radiation effects , Humans , Inflammation/drug therapy , Leukocytes/immunology , Leukocytes/metabolism , Lipoxins/pharmacology , Male , Middle Aged , Neutrophils/drug effects , Receptors, Chemokine/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled , Receptors, Lipoxin/metabolism , Skin/drug effects , Skin/pathology , Volunteers , Young Adult
16.
Einstein (Sao Paulo) ; 15(3): 256-261, 2017.
Article in English, Portuguese | MEDLINE | ID: mdl-29091144

ABSTRACT

OBJECTIVE: To describe e compare the specificity of IgA antibodies against bacteria extract of Klebsiella pneumoniae , Staphylococcus aureus , Escherichia coli , and Salmonella enteritidis . METHODS: Colostrum samples were aseptically collected in the first 12 hours after C-section delivery. The specificity of IgA against bacteria extracts was analyzed by the Western blot. RESULTS: The findings showed proteins of high molecular weight frequently detectable in the samples. S. aureus was the most frequently found bacterium in the samples (p<0.05). Approximately 93.8, 56.3, 62.5 and 60.4% of samples presented IgA reactive to S. aureus , K. pneumoniae , S. enteritidis, and E. coli, respectively. Roughly 40% of samples showed no IgA reactive to K. pneumoniae, S. enteritidis and E. coli . CONCLUSION: Clinical evidence of the importance of breastfeeding for the immune protection of neonates was consistent with the observed immunological findings, since most samples showed IgA reactive against the species tested. The application and development of immunotherapies during pregnancy, focused on frequently detected antigens, could be an important tool to enhance the presence of IgA in colostrum.


Subject(s)
Antibodies, Bacterial/analysis , Colostrum/immunology , Escherichia coli/immunology , Immunoglobulin A, Secretory/analysis , Klebsiella pneumoniae/immunology , Salmonella enteritidis/immunology , Staphylococcus aureus/immunology , Antibodies, Bacterial/immunology , Blotting, Western , Female , Humans , Immunoglobulin A, Secretory/immunology , Infant, Newborn , Sensitivity and Specificity
17.
Nutr Res ; 46: 88-95, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29132841

ABSTRACT

This study hypothesized that the predominant strawberry anthocyanin, pelargonidin-3-O-glucoside (Pg-3-glc), and 3 of its plasma metabolites (4-hydroxybenzoic acid, protocatechuic acid, and phloroglucinaldehyde [PGA]) would affect phagocytosis, oxidative burst, and the production of selected pro- and anti-inflammatory cytokines in a whole blood culture model. For the assessment of phagocytosis and oxidative burst activity of monocytes and neutrophils, whole blood was preincubated in the presence or absence of the test compounds at concentrations up to 5 µmol/L, followed by analysis of phagocytic and oxidative burst activity using commercially available test kits. For the cytokine analysis, diluted whole blood was stimulated with lipopolysaccharide in the presence or absence of the test compounds at concentrations up to 5 µmol/L. Concentrations of selected cytokines (tumor necrosis factor-α, interleukin [IL]-1ß, IL-6, IL-8, and IL-10) were determined using a cytometric bead array kit. There were no effects of any of the test compounds on phagocytosis of opsonized or nonopsonized Escherichia coli or on oxidative burst activity. Pg-3-glc and PGA at 0.08 µmol/L increased the concentration of IL-10 (P<.01 and P<.001, respectively), but there was no effect on tumor necrosis factor-α, IL-1ß, IL-6, and IL-8, and there were no effects of the other compounds. In conclusion, this study demonstrated a lack of effect of these compounds on the opsonization, engulfment, and subsequent destruction of bacteria. Pg-3-glc and PGA, at physiologically relevant concentrations, had anti-inflammatory properties; however, effects were modest, only observed at the lowest dose tested and limited to IL-10.


Subject(s)
Anthocyanins/metabolism , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Benzaldehydes/metabolism , Hydroxybenzoates/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Adult , Aged , Cells, Cultured , Cytokines/metabolism , Dietary Supplements , Escherichia coli/immunology , Female , Humans , Lipopolysaccharides/toxicity , Male , Middle Aged , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Osmolar Concentration , Phagocytosis/drug effects , Pilot Projects , Reproducibility of Results , Respiratory Burst/drug effects
18.
Einstein (Säo Paulo) ; 15(3): 256-261, July-Sept. 2017. tab, graf
Article in English | LILACS | ID: biblio-891403

ABSTRACT

ABSTRACT Objective To describe e compare the specificity of IgA antibodies against bacteria extract of Klebsiella pneumoniae , Staphylococcus aureus , Escherichia coli , and Salmonella enteritidis . Methods Colostrum samples were aseptically collected in the first 12 hours after C-section delivery. The specificity of IgA against bacteria extracts was analyzed by the Western blot. Results The findings showed proteins of high molecular weight frequently detectable in the samples. S. aureus was the most frequently found bacterium in the samples (p<0.05). Approximately 93.8, 56.3, 62.5 and 60.4% of samples presented IgA reactive to S. aureus , K. pneumoniae , S. enteritidis, and E. coli, respectively. Roughly 40% of samples showed no IgA reactive to K. pneumoniae, S. enteritidis and E. coli . Conclusion Clinical evidence of the importance of breastfeeding for the immune protection of neonates was consistent with the observed immunological findings, since most samples showed IgA reactive against the species tested. The application and development of immunotherapies during pregnancy, focused on frequently detected antigens, could be an important tool to enhance the presence of IgA in colostrum.


RESUMO Objetivo Descrever e comparar a especificidade de anticorpos IgA de amostras de colostro contra extratos bacterianos de Klebsiella pneumoniae , Staphylococcus aureus , Escherichia coli e Salmonella enteritidis . Métodos As amostras de colostro foram coletadas assepticamente nas primeiras 12 horas após o nascimento por cesariana. A especificidade de IgA contra extratos de bactérias foi analisada por Western blot. Resultados Os achados mostraram proteínas de alto peso molecular frequentemente detectáveis nas amostras. S. aureus foi a bactéria mais encontrada nas amostras (p<0,05). Cerca de 93,8, 56,3, 62,5 e 60,4% das amostras apresentaram IgA reativa a S. aureus , K. pneumoniae , S. enteritidis e E. coli , respectivamente. Aproximadamente 40% das amostras não apresentaram IgA reativa contra K. pneumoniae , S. enteritidis e E. coli. Conclusão A evidência clínica da importância da amamentação para proteção imunológica ao recém-nascido foi consistente com os achados imunológicos observados, uma vez que a maioria das amostras mostrou IgA reativa contra as espécies testadas. A aplicação e o desenvolvimento de imunoterapias durante a gestação, focada nos antígenos frequentemente detectados, poderiam ser importantes instrumentos para aumentar a presença de IgA no colostro.


Subject(s)
Humans , Female , Infant, Newborn , Salmonella enteritidis/immunology , Staphylococcus aureus/immunology , Immunoglobulin A, Secretory/analysis , Colostrum/immunology , Escherichia coli/immunology , Klebsiella pneumoniae/immunology , Antibodies, Bacterial/analysis , Immunoglobulin A, Secretory/immunology , Blotting, Western , Sensitivity and Specificity , Antibodies, Bacterial/immunology
19.
Pharmacol Res ; 119: 303-312, 2017 05.
Article in English | MEDLINE | ID: mdl-28249816

ABSTRACT

In obesity, gut microbiota LPS may translocate into the blood stream and then contribute to adipose tissue inflammation and oxidative stress, leading to insulin resistance. A causal link between periodontal infection, obesity and type 2 diabetes has also been suggested. We evaluated the ability of polyphenols from Antirhea borbonica medicinal plant to improve the inflammatory and redox status of 3T3-L1 adipocytes exposed to LPS of Porphyromonas gingivalis periodontopathogen or Escherichia coli enterobacteria. Our results show that LPS enhanced the production of Toll-like receptor-dependent MyD88 and NFκB signaling factors as well as IL-6, MCP-1, PAI-1 and resistin. Plant polyphenols reduced LPS pro-inflammatory action. Concomitantly, polyphenols increased the production of adiponectin and PPARγ, known as key anti-inflammatory and insulin-sensitizing mediators. Moreover, both LPS increased intracellular ROS levels and the expression of genes encoding ROS-producing enzymes including NOX2, NOX4 and iNOS. Plant polyphenols reversed these effects and up-regulated MnSOD and catalase antioxidant enzyme gene expression. Noticeably, preconditioning of cells with caffeic acid, chlorogenic acid or kaempferol identified among A. borbonica major polyphenols, led to similar protective properties. Altogether, these findings demonstrate the anti-inflammatory and antioxidant effects of A. borbonica polyphenols on adipocytes, in response to P. gingivalis or E. coli LPS. It will be of major interest to assess A. borbonica polyphenol benefits against obesity-related metabolic disorders such as insulin resistance in vivo.


Subject(s)
Adipocytes/drug effects , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Escherichia coli/immunology , Lipopolysaccharides/immunology , Polyphenols/pharmacology , Porphyromonas gingivalis/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/microbiology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Mice , Oxidative Stress/drug effects , Plants, Medicinal/chemistry , Polyphenols/chemistry , Polyphenols/isolation & purification , Rubiaceae/chemistry
20.
BMC Complement Altern Med ; 16(1): 467, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27846826

ABSTRACT

BACKGROUND: Acute systemic inflammatory response syndrome arising from infection can lead to multiple organ failure and death, with greater susceptibility occurring in immunocompromised individuals. Moreover, sub-acute chronic inflammation is a contributor to the pathology of diverse degenerative diseases (Parkinson's disease, Alzheimer's disease and arthritis). Given the known limitations in Western medicine to treat a broad range of inflammatory related illness as well as the emergence of antibiotic resistance, there is a renewed interest in complementary and alternative medicines (CAMs) to achieve these means. METHODS: A high throughput (HTP) screening of >1400 commonly sold natural products (bulk herbs, cooking spices, teas, leaves, supplement components, nutraceutical food components, fruit and vegetables, rinds, seeds, polyphenolics etc.) was conducted to elucidate anti-inflammatory substances in lipopolysaccharide (LPS) (E. coli serotype O111:B4) monocytes: RAW 264.7 macrophages [peripheral], BV-2 microglia [brain]) relative to hydrocortisone, dexamethasone and L-N6-(1Iminoethyl)lysine (L-NIL). HTP evaluation was also carried out for lethal kill curves against E.coli 0157:H7 1x106 CFU/mL relative to penicillin. Validation studies were performed to assess cytokine profiling using antibody arrays. Findings were corroborated by independent ELISAs and NO2-/iNOS expression quantified using the Griess Reagent and immunocytochemistry, respectively. For robust screening, we developed an in-vitro efficacy paradigm to ensure anti-inflammatory parameters were observed independent of cytotoxicity. This caution was taken given that many plants exert tumoricidal and anti-inflammatory effects at close range through similar signaling pathways, which could lead to false positives. RESULTS: The data show that activated BV-2 microglia cells (+ LPS 1µg/ml) release >10-fold greater IL-6, MIP1/2, RANTES and nitric oxide (NO2-), where RAW 264.7 macrophages (+ LPS 1µg/ml) produced > 10-fold rise in sTNFR2, MCP-1, IL-6, GCSF, RANTES and NO2-. Data validation studies establish hydrocortisone and dexamethasone as suppressing multiple pro-inflammatory processes, where L-NIL suppressed NO2-, but had no effect on iNOS expression or IL-6. The screening results demonstrate relative few valid hits with anti-inflammatory effects at < 250µg/ml for the following: Bay Leaf (Laurus nobilis), Elecampagne Root (Inula helenium), Tansy (Tanacetum vulgare),Yerba (Eriodictyon californicum) and Centipeda (Centipeda minima), Ashwagandha (Withania somnifera), Feverfew (Tanacetum parthenium), Rosemary (Rosmarinus officinalis), Turmeric Root (Curcuma Longa), Osha Root (Ligusticum porteri), Green Tea (Camellia sinensis) and constituents: cardamonin, apigenin, quercetin, biochanin A, eupatorin, (-)-epigallocatechin gallate (EGCG) and butein. Natural products lethal against [E. coli 0157:H7] where the LC50 < 100 µg/ml included bioactive silver hydrosol-Argentyn 23, green tea (its constituents EGCG > Polyphenon 60 > (-)-Gallocatechin > Epicatechin > (+)-Catechin), Grapeseed Extract (Vitis vinifera), Chinese Gallnut (its constituents gallic acid > caffeic acid) and gallic acid containing plants such as Babul Chall Bark (Acacia Arabica), Arjun (Terminalia Arjuna) and Bayberry Root Bark (Morella Cerifera). CONCLUSIONS: These findings emphasize and validate the previous work of others and identify the most effective CAM anti-inflammatory, antibacterial compounds using these models. Future work will be required to evaluate potential combination strategies for long-term use to prevent chronic inflammation and possibly lower the risk of sepsis in immunocompromised at risk populations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Escherichia coli/drug effects , Macrophages/drug effects , Microglia/drug effects , Sepsis/immunology , Animals , Drug Evaluation, Preclinical , Escherichia coli/immunology , High-Throughput Screening Assays , Humans , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Macrophages/immunology , Mice , Microglia/immunology , RAW 264.7 Cells , Sepsis/drug therapy , Sepsis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL