Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Immunol ; 12: 745332, 2021.
Article in English | MEDLINE | ID: mdl-34671359

ABSTRACT

The induction of trained immunity represents an emerging concept defined as the ability of innate immune cells to acquire a memory phenotype, which is a typical hallmark of the adaptive response. Key points modulated during the establishment of trained immunity include epigenetic, metabolic and functional changes in different innate-immune and non-immune cells. Regarding to epigenetic changes, it has been described that long non-coding RNAs (LncRNAs) act as molecular scaffolds to allow the assembly of chromatin-remodeling complexes that catalyze epigenetic changes on chromatin. On the other hand, relevant metabolic changes that occur during this process include increased glycolytic rate and the accumulation of metabolites from the tricarboxylic acid (TCA) cycle, which subsequently regulate the activity of histone-modifying enzymes that ultimately drive epigenetic changes. Functional consequences of established trained immunity include enhanced cytokine production, increased antigen presentation and augmented antimicrobial responses. In this article, we will discuss the current knowledge regarding the ability of different cell subsets to acquire a trained immune phenotype and the molecular mechanisms involved in triggering such a response. This knowledge will be helpful for the development of broad-spectrum therapies against infectious diseases based on the modulation of epigenetic and metabolic cues regulating the development of trained immunity.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Cellular , Immunity, Innate/immunology , Immunologic Memory/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Adaptive Immunity/physiology , Animals , BCG Vaccine/immunology , Bronchi/cytology , Bronchi/immunology , Cytokines/physiology , Energy Metabolism , Epigenesis, Genetic , Epithelial Cells/immunology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Hematopoietic Stem Cells/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/physiology , Immunity, Innate/genetics , Immunity, Innate/physiology , Immunologic Memory/genetics , Immunologic Memory/physiology , Lymphocytes/immunology , Mice , Myeloid Cells/immunology , NAD/physiology , Skin/cytology , Skin/immunology
2.
Cell Mol Gastroenterol Hepatol ; 12(2): 489-506, 2021.
Article in English | MEDLINE | ID: mdl-33864900

ABSTRACT

BACKGROUND AND AIMS: CD4+ T cells constitute central players in inflammatory bowel diseases (IBDs), driving inflammation in the gut mucosa. Current evidence indicates that CCR9 and the integrin α4ß7 are necessary and sufficient to imprint colonic homing on CD4+ T cells upon inflammation. Interestingly, dopaminergic signaling has been previously involved in leukocyte homing. Despite dopamine levels are strongly reduced in the inflamed gut mucosa, the role of dopamine in the gut homing of T cells remains unknown. Here, we study how dopaminergic signaling affects T cells upon gut inflammation. METHODS: Gut inflammation was induced by transfer of naïve T cells into Rag1-/- mice or by administration of dextran sodium sulfate. T cell migration and differentiation were evaluated by adoptive transfer of congenic lymphocytes followed by flow cytometry analysis. Protein interaction was studied by bioluminescence resonance energy transfer analysis, bimolecular fluorescence complementation, and in situ proximity ligation assays. RESULTS: We show the surface receptor providing colonic tropism to effector CD4+ T cells upon inflammation is not CCR9 but the complex formed by CCR9 and the dopamine receptor D5 (DRD5). Assembly of the heteromeric complex was demonstrated in vitro and in vivo using samples from mouse and human origin. The CCR9:DRD5 heteroreceptor was upregulated in the intestinal mucosa of IBD patients. Signaling assays confirmed that complexes behave differently than individual receptors. Remarkably, the disruption of CCR9:DRD5 assembly attenuated the recruitment of CD4+ T cells into the colonic mucosa. CONCLUSIONS: Our findings describe a key homing receptor involved in gut inflammation and introduce a new cell surface module in immune cells: macromolecular complexes formed by G protein-coupled receptors integrating the sensing of multiple molecular cues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Inflammation/immunology , Protein Multimerization , Receptors, CCR/metabolism , Receptors, Dopamine D5/metabolism , Amino Acid Sequence , Animals , Cell Movement , Cell Proliferation , Colitis/immunology , Colitis/pathology , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Integrin beta1/metabolism , Jurkat Cells , MAP Kinase Signaling System , Mice, Inbred C57BL , Models, Biological , Peptides/chemistry , Phosphorylation , Receptors, CCR/deficiency , Receptors, Dopamine D5/deficiency , Signal Transduction , Tropism
4.
Food Res Int ; 136: 109577, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32846611

ABSTRACT

The year 2020 will be remembered by a never before seen, at least by our generation, global pandemic of COVID-19. While a desperate search for effective vaccines or drug therapies is on the run, nutritional strategies to promote immunity against SARS-CoV-2, are being discussed. Certain fermented foods and probiotics may deliver viable microbes with the potential to promote gut immunity. Prebiotics, on their side, may enhance gut immunity by selectively stimulating certain resident microbes in the gut. Different levels of evidence support the use of fermented foods, probiotics and prebiotics to promote gut and lungs immunity. Without being a promise of efficacy against COVID-19, incorporating them into the diet may help to low down gut inflammation and to enhance mucosal immunity, to possibly better face the infection by contributing to diminishing the severity or the duration of infection episodes.


Subject(s)
Coronavirus Infections/therapy , Fermented Foods , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammation , Pneumonia, Viral/therapy , Prebiotics , Probiotics , Betacoronavirus , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/microbiology , Coronavirus Infections/virology , Diet , Gastrointestinal Tract/immunology , Humans , Inflammation/etiology , Inflammation/microbiology , Inflammation/prevention & control , Inflammation/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/microbiology , Pneumonia, Viral/virology , SARS-CoV-2
5.
Article in English | MEDLINE | ID: mdl-32368985

ABSTRACT

BACKGROUND: Triple immunosuppressive therapy is associated with several gastrointestinal disorders. The aim of this study was to investigate the effects induced by the triple immunosuppressive therapy on the gastrointestinal tract of rats. METHODS: Male Wistar rats were randomly assigned into three experimental groups: Control: filtered water; TAC + MPS + PRED: treated with Tacrolimus plus Mycophenolate Sodium plus Prednisone; and CSA + AZA + PRED: treated with Cyclosporine plus Azathioprine plus Prednisone. The treatment was done for 14 days by gavage. Gastric emptying and contractility were evaluated by the Alternating Current Biosusceptometry (ACB) and Electrogastrography (EGG). Histological, biochemical and hematological analyses were also performed. RESULTS: Gastric emptying time was slower in the CSA + AZA + PRED group in comparison with control (p<0.01) and TAC + MPS + PRED groups (p<0.001). Animals treated with TAC + MPS + PRED showed accelerated gastric emptying (p<0.05) compared to control. The amplitude of gastric contractions in both immunosuppressed groups was higher than observed in the control. The frequency of gastric contractions for the CSA + AZA + PRED group was also increased (p<0.01). Results obtained by EGG were similar to those recorded with the ACB. The thickness of the circular layer from stomach muscle decreased in both immunosuppressed groups, while the longitudinal layer was reduced only in the CSA + AZA + PRED group. CONCLUSION: Triple immunosuppressive therapy alters gastric motility, compromises the muscular layers and the association between CSA, AZA, and PRED provokes the major alterations in the structure and gastric function. Specific gastrointestinal side effects resulting from different immunosuppressive therapies still need to be elucidated in order to provide more effective and personalized therapy for patients.


Subject(s)
Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/immunology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Animals , Azathioprine/administration & dosage , Azathioprine/adverse effects , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Drug Therapy, Combination , Gastric Emptying/drug effects , Gastric Emptying/immunology , Gastrointestinal Diseases/pathology , Male , Rats , Rats, Wistar , Tacrolimus/administration & dosage , Tacrolimus/adverse effects
6.
Food Res Int ; 125: 108567, 2019 11.
Article in English | MEDLINE | ID: mdl-31554075

ABSTRACT

This randomized, double-blind, parallel and placebo-controlled study aimed to evaluate the effect of Bacillus coagulans GBI-30, 6086® probiotic (GanedenBC30®) against upper respiratory tract infections (URTI) and gastrointestinal tract infections (GITI) in eighty healthy school-aged children (6-8 years old). The participants received daily a sachet containing either GanedenBC30 (1 × 109 colony-forming units) or placebo (maltodextrin) for three months. GanedenBC30 significantly decreased the incidence of URTI symptoms including nasal congestion, bloody nasal mucus, itchy nose, and hoarseness. The duration of the URTI-associated symptoms of hoarseness, headache, red eyes, and fatigue was also decreased. GanedenBC30 supplementation also significantly reduced the incidence rate of flatulence. These beneficial effects were associated with the modulation of serum TNFα, CD163, G-CSF, ICAM-1, IL-6, IL-8, MCP-2, RAGE, uPAR, and PF4. Therefore, probiotic B. coagulans GBI-30, 6086 modulated immune-related proteins in healthy children, decreasing several URTI and GITI symptoms, thus, this functional ingredient may contribute to a healthier lifestyle.


Subject(s)
Bacillus coagulans/immunology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/prevention & control , Probiotics/pharmacology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Child , Double-Blind Method , Female , Gastrointestinal Diseases/immunology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Humans , Incidence , Male , Mexico/epidemiology , Respiratory System/drug effects , Respiratory System/immunology , Respiratory Tract Infections/immunology , Severity of Illness Index , Time
7.
Eur J Immunol ; 49(11): 2095-2102, 2019 11.
Article in English | MEDLINE | ID: mdl-31334839

ABSTRACT

There is increasing evidence of the relevant connection and regulation between the gut and skin immune axis. In fact, oral administration of lipoteichoic acid (LTA) from Lactobacillus rhamnosus GG (LGG) prevents the development of UV-induced skin tumors in chronically exposed mice. Here we aim to evaluate whether this LTA is able to revert UV-induced immunosuppression as a mechanism involved in its anti-tumor effect and whether it has an immunotherapeutic effect against cutaneous squamous cell carcinoma. Using a mouse model of contact hypersensitivity, we demonstrate that LTA overcomes UV-induced skin immunosuppression. This effect was in part achieved by modulating the phenotype of lymph node resident dendritic cells (DC) and the homing of skin migratory DC. Importantly, oral LTA reduced significantly the growth of established skin tumors once UV radiation was discontinued, demonstrating that it has a therapeutic, besides the already demonstrated preventive antitumor effect. The data presented here strongly indicates that oral administration of LTA represents a promising immunotherapeutic approach for different conditions in which the skin immune system is compromised.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Lacticaseibacillus rhamnosus/chemistry , Lipopolysaccharides/pharmacology , Skin Neoplasms/drug therapy , Teichoic Acids/pharmacology , Ultraviolet Rays/adverse effects , Administration, Oral , Animals , Antineoplastic Agents/isolation & purification , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Movement/drug effects , Cell Movement/immunology , Cell Movement/radiation effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/radiation effects , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/radiation effects , Lipopolysaccharides/isolation & purification , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/radiation effects , Mice , Mice, Inbred C57BL , Skin/drug effects , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Teichoic Acids/isolation & purification
8.
Rheumatology (Oxford) ; 58(11): 1985-1990, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31056685

ABSTRACT

OBJECTIVES: Changes in the intestinal microbiota have been associated with the pathogenesis of SSc. Probiotics act by modulating the microbiome and the immune response. This study aimed to evaluate the efficacy of probiotics on gastrointestinal (GI) symptoms and immune responses in SSc patients. METHODS: Patients with SSc with a moderate-severe total score on the University of California Los Angeles Scleroderma Clinical Trials Consortium Gastrointestinal Tract 2.0 (UCLA GIT 2.0) instrument were randomly assigned to receive a daily dose of probiotics (Lactobacillus paracasei, Lactobacillus rhamnosus, Lactobacillus acidophillus and Bifidobacterium lactis, 109 colony-forming units per capsule) or placebo for 8 weeks. The primary endpoint was improvement in the UCLA GIT 2.0 total score after 8 weeks. Secondary outcomes included changes in Th1, Th2, Th17 and regulatory T cell circulating levels and in the HAQ Disability Index (HAQ-DI) score. Parameters were assessed at baseline and after 4 and 8 weeks of treatment. RESULTS: A total of 73 patients were randomized to receive probiotics (n = 37) or placebo (n = 36). After 8 weeks, there was no difference in the UCLA GIT 2.0 score between the two groups. At week 8, the probiotic group showed a significant decrease in the proportion of Th17 cells compared with placebo (P = 0.003). There was no difference in the proportion of Th1, Th2 and regulatory T cells or in the HAQ-DI score between the groups. CONCLUSION: Probiotics did not improve GI symptoms in SSc patients. The reduction in Th17 cell levels suggests an immunomodulatory effect of probiotics on SSc. TRIAL REGISTRATION: ClinicalTrials.gov (http://clinicaltrials.gov), NCT02302352.


Subject(s)
Gastrointestinal Diseases/therapy , Probiotics/therapeutic use , Scleroderma, Systemic/immunology , Scleroderma, Systemic/microbiology , Adult , Disability Evaluation , Double-Blind Method , Female , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Humans , Male , Middle Aged , Scleroderma, Systemic/complications , Severity of Illness Index , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Treatment Outcome
9.
Front Immunol ; 10: 239, 2019.
Article in English | MEDLINE | ID: mdl-30828335

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder affecting mainly the dopaminergic neurons of the nigrostriatal pathway, a neuronal circuit involved in the control of movements, thereby the main manifestations correspond to motor impairments. The major molecular hallmark of this disease corresponds to the presence of pathological protein inclusions called Lewy bodies in the midbrain of patients, which have been extensively associated with neurotoxic effects. Importantly, different research groups have demonstrated that CD4+ T-cells infiltrate into the substantia nigra of PD patients and animal models. Moreover, several studies have consistently demonstrated that T-cell deficiency results in a strong attenuation of dopaminergic neurodegeneration in animal models of PD, thus indicating a key role of adaptive immunity in the neurodegenerative process. Recent evidence has shown that CD4+ T-cell response involved in PD patients is directed to oxidised forms of α-synuclein, one of the main constituents of Lewy bodies. On the other hand, most PD patients present a number of non-motor manifestations. Among non-motor manifestations, gastrointestinal dysfunctions result especially important as potential early biomarkers of PD, since they are ubiquitously found among confirmed patients and occur much earlier than motor symptoms. These gastrointestinal dysfunctions include constipation and inflammation of the gut mucosa and the most distinctive pathologic features associated are the loss of neurons of the enteric nervous system and the generation of Lewy bodies in the gut. Moreover, emerging evidence has recently shown a pivotal role of gut microbiota in triggering the development of PD in genetically predisposed individuals. Of note, PD has been positively correlated with inflammatory bowel diseases, a group of disorders involving a T-cell driven inflammation of gut mucosa, which is strongly dependent in the composition of gut microbiota. Here we raised the hypothesis that T-cell driven inflammation, which mediates dopaminergic neurodegeneration in PD, is triggered in the gut mucosa. Accordingly, we discuss how structural components of commensal bacteria or how different mediators produced by gut-microbiota, including short-chain fatty acids and dopamine, may affect the behaviour of T-cells, triggering the development of T-cell responses against Lewy bodies, initially confined to the gut mucosa but later extended to the brain.


Subject(s)
Brain/metabolism , Dopaminergic Neurons/pathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Inflammation/immunology , Parkinson Disease/immunology , T-Lymphocytes/immunology , Brain/pathology , Humans , Immunity, Cellular , Neuroimmunomodulation
11.
Fish Shellfish Immunol ; 75: 139-148, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29421586

ABSTRACT

Eleginops maclovinus is an endemic fish to Chile that lives in proximity to salmonid culture centers, feeding off of uneaten pellet and salmonid feces. Occurring in the natural environment, this interaction between native and farmed fish could result in the horizontal transmission of pathogens affecting the aquaculture industry. The aim of this study was to evaluate the innate and adaptive immune responses of E. maclovinus challenged with P. salmonis. Treatment injections (in duplicate) were as follows: control (100 µL of culture medium), wild type LF-89 strain (100 µL, 1 × 108 live bacteria), and antibiotic resistant strain Austral-005 (100 µL, 1 × 108 live bacteria). The fish were sampled at various time-points during the 35-day experimental period. The gene expression of TLRs (1, 5, and 8), NLRCs (3 and 5), C3, IL-1ß, MHCII, and IgMs were significantly modulated during the experimental period in both the spleen and gut (excepting TLR1 and TLR8 spleen expressions), with tissue-specific expression profiles and punctual differences between the injected strains. Anti-P. salmonis antibodies increased in E. maclovinus serum from day 14-28 for the LF-89 strain and from day 14-35 for the Austral-005 strain. These results suggest temporal activation of the innate and adaptive immune responses in E. maclovinus tissues when injected by distinct P. salmonis strains. The Austral-005 strain did not always cause the greatest increases/decreases in the number of transcripts, so the magnitude of the observed immune response (mRNA) may not be related to antibiotic resistance. This is the first immunological study to relate a pathogen widely studied in salmonids with a native fish.


Subject(s)
Adaptive Immunity , Fish Diseases/immunology , Immunity, Innate , Perciformes/immunology , Piscirickettsia/physiology , Piscirickettsiaceae Infections/veterinary , Animals , Antarctic Regions , Chile , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Piscirickettsia/genetics , Piscirickettsiaceae Infections/immunology , Random Allocation , Spleen/immunology , Spleen/microbiology , Time Factors
13.
Tissue Barriers ; 5(4): e1373208, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28956703

ABSTRACT

The gastrointestinal (GI) tract is considered the largest immunological organ in the body having a central role in regulating immune homeostasis. Contrary to earlier belief, the intestinal epithelial barrier is not a static physical barrier but rather strongly interacts with the gut microbiome and cells of the immune system. This intense communication between epithelial cells, immune cells and microbiome will shape specific immune responses to antigens, balancing tolerance and effector immune functions. Recent studies indicate that composition of the gut microbiome affects immune system development and modulates immune mediators, which in turn affect the intestinal barrier. Moreover, dysbiosis may favor intestinal barrier disruption and could be related to increased susceptibility to certain diseases. This review will be focused on the development of the intestinal barrier and its function in host immune defense and how gut microbiome composition throughout life can affect this role.


Subject(s)
Adaptive Immunity/physiology , Aging/immunology , Aging/metabolism , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Humans , Immune Tolerance/physiology
14.
J Med Microbiol ; 65(12): 1347-1362, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27902422

ABSTRACT

Cancer is defined as an uncontrolled proliferation of malignant cells in a host and it is one of the main causes of death worldwide. Genetic and environmental factors play an important role in its development, and the involvement of microbial communities has also recently been recognized. The close relationship that characterizes the colonization by human commensal communities involves health risks, particularly when the homeostasis is disturbed. It has been hypothesized that this process may lead to cancer by modulating the inflammatory response of the host, by the production of carcinogenic metabolic products or by the production of toxins, which disrupt the cell cycle. The metabolic effects of the intestinal microbiota have been studied in greater detail in the gastrointestinal tract, and it has been recognized that microbial communities of other body surfaces can cause effects either locally or at a distance. In vitro and in vivo studies have allowed the characterization of the microbiota and the establishment of a cause and effect relationship with some types of cancer. Nevertheless, despite the results, representative studies are necessary to validate the findings and definitively establish the role of microbiota in cancer development in order to open the possibility of promising therapeutic and diagnostic applications. Thus, the aims of this review are to briefly examine the available evidence, and to analyse the mechanisms described for pancreatic, lung, colorectal cancer , oral squamous cell carcinoma and hepatocellular carcinoma and the impact of the current knowledge about the effects of the microbiota on carcinogenesis.


Subject(s)
Carcinogenesis , Dysbiosis , Gastrointestinal Microbiome/physiology , Animals , Colorectal Neoplasms/etiology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Host-Pathogen Interactions , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/microbiology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/microbiology , Probiotics
15.
Mediators Inflamm ; 2016: 1363818, 2016.
Article in English | MEDLINE | ID: mdl-27471349

ABSTRACT

Intestinal immunity is finely regulated by several concomitant and overlapping mechanisms, in order to efficiently sense external stimuli and mount an adequate response of either tolerance or defense. In this context, a complex interplay between immune and nonimmune cells is responsible for the maintenance of normal homeostasis. However, in certain conditions, the disruption of such an intricate network may result in intestinal inflammation, including inflammatory bowel disease (IBD). IBD is believed to result from a combination of genetic and environmental factors acting in concert with an inappropriate immune response, which in turn interacts with nonimmune cells, including nervous system components. Currently, evidence shows that the interaction between the immune and the nervous system is bidirectional and plays a critical role in the regulation of intestinal inflammation. Recently, the maintenance of intestinal homeostasis has been shown to be under the reciprocal control of the microbiota by immune mechanisms, whereas intestinal microorganisms can modulate mucosal immunity. Therefore, in addition to presenting the mechanisms underlying the interaction between immune and nervous systems in the gut, here we discuss the role of the microbiota also in the regulation of neuroimmune crosstalk involved in intestinal homeostasis and inflammation, with potential implications to IBD pathogenesis.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Neuroimmunomodulation/physiology , Animals , Gastrointestinal Tract/pathology , Homeostasis , Humans , Inflammatory Bowel Diseases/pathology
16.
J Pediatr ; 173 Suppl: S16-28, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27234407

ABSTRACT

The early postnatal period is a critical time for gastrointestinal (GI) and immune development. Neonates fed mother's milk have more rapid GI and immune development than fed-formula infants. In addition, clinical and epidemiologic data provide strong evidence that breastfeeding reduces the incidence and/or severity of infectious diseases. Lactoferrin is a 77 kDa, iron-binding glycoprotein that is present at high concentration in human milk compared with bovine milk and infant formula. It is a multifunctional protein that mediates many of the physiological processes in which breastfed infants have advantages over their formula-fed peers, including promoting GI and immune development, protection from infections, and improved cognitive development. Feeding bovine lactoferrin or recombinant human lactoferrin was well tolerated and stimulated intestinal cell proliferation and increased villus length and crypt depth in piglets. Lactoferrin also influenced both systemic and GI immune development by stimulating a balanced T-helper-1/T-helper-2 cytokine immune response. Further, there was a tendency for immune cells to secrete more anti-inflammatory cytokines in an unstimulated state, while being primed for a robust pro-inflammatory response when presented with a bacterial trigger in piglets fed lactoferrin. These findings support clinical studies demonstrating benefits of dietary lactoferrin in the prevention of infections, late onset sepsis, and necrotizing enterocolitis.


Subject(s)
Gastrointestinal Tract/growth & development , Immune System/growth & development , Infant Nutritional Physiological Phenomena , Lactoferrin/physiology , Milk/physiology , Animals , Breast Feeding , Cattle , Enterocolitis, Necrotizing/prevention & control , Gastrointestinal Tract/immunology , Humans , Infant , Infant Formula , Infant, Newborn , Lactoferrin/chemistry , Milk/chemistry , Models, Animal , Sepsis/prevention & control , Swine , Translational Research, Biomedical
17.
J Pediatr ; 173 Suppl: S43-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27234411

ABSTRACT

To discuss the potential clinical benefits of lactoferrin in preterm and term infants, as well as in young children and to review information on the burden of neonatal sepsis. Current evidence on the mechanisms that explain the role of human milk in the neonatal and infant anti-infective responses will be briefly reviewed and preclinical research data on the potential mechanisms of action by which lactoferrin may impact infant gut health, gut immune development and functions, including the lactoferrin effects on the neonatal microbiome, will be examined. Finally, updated translational research on lactoferrin will be presented and discussed and the current evidence from prospective randomized controlled trials in neonates, infants, and toddlers will be analyzed. These randomized controlled trials demonstrate that lactoferrin has a clinically significant impact on feeding, the microbiome, and clinical outcomes in neonates and infants.


Subject(s)
Anti-Infective Agents/therapeutic use , Candidiasis/prevention & control , Enterocolitis, Necrotizing/prevention & control , Gastrointestinal Tract/immunology , Lactoferrin/therapeutic use , Milk, Human/immunology , Sepsis/prevention & control , Candidiasis/immunology , Child, Preschool , Enterocolitis, Necrotizing/immunology , Humans , Infant , Infant Nutritional Physiological Phenomena/immunology , Infant, Newborn , Intensive Care, Neonatal/methods , Lactoferrin/immunology , Microbiota/immunology , Milk, Human/chemistry , Sepsis/immunology
18.
J Pediatr ; 173 Suppl: S53-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27234412

ABSTRACT

Sphingomyelin (SM), glycosphingolipids, and gangliosides are important polar lipids in the milk fat globule membrane but are not found in standard milk replacement formulas. Because digestion and absorption of SM and glycosphingolipids generate the bioactive metabolites ceramide, sphingosine, and sphingosine-1-phosphate (S1P), and because intact gangliosides may have beneficial effects in the gut, this may be important for gut integrity and immune maturation in the neonate. The brush border enzymes that hydrolyze milk SM, alkaline sphingomyelinase (nucleotide phosphodiesterase pyrophosphatase 7), and neutral ceramidase are expressed at birth in both term and preterm infants. Released sphingosine is absorbed, phosphorylated to S1P, and converted to palmitic acid via S1P-lyase in the gut mucosa. Hypothetically, S1P also may be released from absorptive cells and exert important paracrine actions favoring epithelial integrity and renewal, as well as immune function, including secretory IgA production and migration of T lymphocyte subpopulations. Gluco-, galacto-, and lactosylceramide are hydrolyzed to ceramide by lactase-phlorizin hydrolase, which also hydrolyzes lactose. Gangliosides may adhere to the brush border and is internalized, modified, and possibly transported into blood, and may exert protective functions by their interactions with bacteria, bacterial toxins, and the brush border.


Subject(s)
Gastrointestinal Tract/immunology , Infant Nutritional Physiological Phenomena/immunology , Milk, Human/immunology , Sphingolipids/immunology , Gastrointestinal Tract/physiology , Humans , Infant , Infant Nutritional Physiological Phenomena/physiology , Infant, Newborn , Lipolysis/physiology , Milk, Human/chemistry , Milk, Human/physiology , Sphingolipids/physiology
19.
Parasit Vectors ; 9: 119, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26931761

ABSTRACT

BACKGROUND: Rhodnius prolixus is a major vector of Trypanosoma cruzi, the causative agent of Chagas disease in Latin America. In natural habitats, these insects are in contact with a variety of bacteria, fungi, virus and parasites that they acquire from both their environments and the blood of their hosts. Microorganism ingestion may trigger the synthesis of humoral immune factors, including antimicrobial peptides (AMPs). The objective of this study was to compare the expression levels of AMPs (defensins and prolixicin) in the different midgut compartments and the fat body of R. prolixus infected with different T. cruzi strains. The T. cruzi Dm 28c clone (TcI) successfully develops whereas Y strain (TcII) does not complete its life- cycle in R. prolixus. The relative AMP gene expressions were evaluated in the insect midgut and fat body infected on different days with the T. cruzi Dm 28c clone and the Y strain. The influence of the antibacterial activity on the intestinal microbiota was taken into account. METHODS: The presence of T. cruzi in the midgut of R. prolixus was analysed by optical microscope. The relative expression of the antimicrobial peptides encoding genes defensin (defA, defB, defC) and prolixicin (prol) was quantified by RT-qPCR. The antimicrobial activity of the AMPs against Staphylococcus aureus, Escherichia coli and Serratia marcescens were evaluated in vitro using turbidimetric tests with haemolymph, anterior and posterior midgut samples. Midgut bacteria were quantified using colony forming unit (CFU) assays and real time quantitative polymerase chain reaction (RT-qPCR). RESULTS: Our results showed that the infection of R. prolixus by the two different T. cruzi strains exhibited different temporal AMP induction profiles in the anterior and posterior midgut. Insects infected with T. cruzi Dm 28c exhibited an increase in defC and prol transcripts and a simultaneous reduction in the midgut cultivable bacteria population, Serratia marcescens and Rhodococcus rhodnii. In contrast, the T. cruzi Y strain neither induced AMP gene expression in the gut nor reduced the number of colony formation units in the anterior midgut. Beside the induction of a local immune response in the midgut after feeding R. prolixus with T. cruzi, a simultaneous systemic response was also detected in the fat body. CONCLUSIONS: R. prolixus AMP gene expressions and the cultivable midgut bacterial microbiota were modulated in distinct patterns, which depend on the T. cruzi genotype used for infection.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Fat Body/immunology , Gene Expression , Insect Vectors , Rhodnius/immunology , Trypanosoma cruzi/immunology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Colony Count, Microbial , Escherichia coli/drug effects , Fat Body/parasitology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/parasitology , Gene Expression Profiling , Microscopy , Real-Time Polymerase Chain Reaction , Rhodnius/genetics , Rhodnius/parasitology , Serratia marcescens/drug effects , Staphylococcus aureus/drug effects , Trypanosoma cruzi/drug effects
20.
Alcohol Clin Exp Res ; 39(8): 1453-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26110492

ABSTRACT

BACKGROUND: Ethanol (EtOH) consumption is able to disturb the ovalbumin (OVA)-oral tolerance induction by interfering on the function of antigen presenting cells (APC), down-regulating dendritic cells (DCs) and macrophages and up-regulating B-lymphocytes and their function, which results in an overall allergic-type immune status. In this study, the potential of a priori administration of Lactococcus lactis (LL) in avoiding loss of oral tolerance in EtOH-treated mice was investigated. METHODS: Female C57BL/6 mice received, by oral route, ad libitum wild-type (WT) LL or heat-shock protein producer (Hsp65) LL for 4 consecutive days. Seven days later, mice were submitted to short-term high-dose EtOH treatment. After 24 hours, stomach, intestine, spleen, mesenteric lymph nodes (mLN) specimens were collected for biomarkers analysis. Following EtOH-treatment protocol, a group of animals underwent single-gavage OVA-tolerance protocol and sera samples collected for antibody analysis. RESULTS: The ingestion of WT LL or Hsp65 LL is able to restore oral tolerance to OVA in EtOH-treated mice, by reducing local and systemic allergic outcomes such as gastric mast cells and gut-interleukin-4, as well as serum IgE. WT LL treatment prevents the decrease of mLN regulatory T cells induced by the EtOH treatment. Moreover, LL treatment preserves APC hierarchy and antigen presentation commitment in EtOH-treated mice, with conserved DC and macrophage activity over B lymphocytes in mLN and preserved macrophage activity over DC and B-cell subsets in the spleen. CONCLUSIONS: The present findings suggest that a priori ingestion of LL preserves essential mechanisms associated with oral tolerance induction that are disturbed by EtOH ingestion. Maintenance of mucosal homeostasis by preserving APC hierarchy and antigen presentation commitment could be associated with T-regulatory subset activities in the gastrointestinal tract.


Subject(s)
Antigen Presentation/immunology , Ethanol/administration & dosage , Gastrointestinal Tract/immunology , Immune Tolerance/immunology , Lactococcus lactis , Administration, Oral , Animals , Antigen Presentation/drug effects , Female , Gastrointestinal Tract/drug effects , Immune Tolerance/drug effects , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL