Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Immunol ; 17(1): 42, 2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27793087

RESUMO

BACKGROUND: The small intestine is a specialized compartment were close interactions take place between host, microbes, food antigens and dietary fatty acids. Dietary fats get absorbed by epithelial cells and processed into a range of lipoprotein particles after which they are basolaterally secreted and collected in the lymphatics. In contrast to the colon, the small intestine is covered only by a thin mucus coat that allows for intimate interactions between host-cells and microbes. Lipoproteins have long been recognized as protective factors in infectious diseases via the neutralization of bacterial toxins like lipopolysaccharides. Much less attention has been given to the potential role of lipoproteins as factors contributing to the maintenance of small intestinal immune homeostasis via modulating bacteria-induced immune responses. RESULTS: Lipoproteins VLDL, LDL and HDL were found to neutralize TLR responses towards specific TLR-ligands or a selection of gram-negative and gram-positive bacteria. Attenuation of TLR2 activity was acute and only slightly improved by longer pre-incubation times of ligands and lipoproteins with no differences between bacterial-lipopeptides or bacteria. In contrast, attenuation of TLR4 responses was only observed after extensive preincubation of lipoproteins and LPS. Preincubation of bacteria and lipoproteins led only to a modest attenuation of TLR4 activity. Moreover, compared to TLR2, TLR4 activity could only be attenuated by lipoproteins over a small ligand dose range. CONCLUSIONS: These results demonstrate the ability of lipoproteins VLDL, LDL and HDL to inhibit TLR responses towards bacterial-ligands and bacteria. Presence of lipoproteins was found to modulate the MAMP-induced cytokine release by primary human monocytes measured as changes in the release of IL-6, TNFα, GM-CSF and IFNγ. Using TLR2 and TLR4-reporter cells, lipoproteins were found to inhibit TLR responses with differences in affinity and kinetics. These data establish a role for lipoproteins as immunoregulatory molecules, attenuating TLR-responses and thereby positively contributing to mucosal homeostasis.


Assuntos
Leucócitos Mononucleares/imunologia , Salmonella typhimurium/imunologia , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Antígenos de Bactérias/imunologia , Apolipoproteínas/imunologia , Citocinas/metabolismo , Células HEK293 , Humanos , Imunização , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/metabolismo , Ativação Linfocitária
2.
J Nutr Biochem ; 26(3): 227-33, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25498760

RESUMO

Inflammatory bowel diseases (IBD) including ulcerative colitis (UC) and Crohn's disease (CD) are chronic relapsing inflammatory disorders of the gastrointestinal tract. The interaction between a disturbed microbial composition, the intestinal mucosal barrier and the mucosal immune system plays an important role in IBD and its chronicity. It has been indicated that due to the altered microbial composition the balance between T regulatory cells (Treg) and T helper cells (Th) 17 is disturbed, leading to an inflammatory state. The present study shows that oral intake of a specific multi fibre mix (MF), designed to match the fibre content of a healthy diet, counteracts IBD-like intestinal inflammation and weight loss in dextran sodium sulphate treated mice. This reduction in inflammation might be brought about, at least in part, by the MF-induced decrease in inflammatory cytokines, increase in IL-10 and the relative increase in Treg cells in the mesenteric lymph nodes (MLN). Moreover, the Treg percentage in the MLN correlates with the percentage of tolerogenic lamina propria derived CD103+RALDH+dendritic cells in the MLN, suggesting that these play a role in the observed effects. In children with CD exclusive enteral nutrition (EEN) is a widely used safe and effective therapy. Optimizing enteral nutritional concepts with the tested fibre mix, know to modulate the gut microbiota composition, SCFA production and inflammatory status (as indicated by the present study) could possibly further improve efficacy in inducing remission.


Assuntos
Colo/imunologia , Modelos Animais de Doenças , Imunomodulação , Doenças Inflamatórias Intestinais/dietoterapia , Mucosa Intestinal/imunologia , Prebióticos , Linfócitos T Reguladores/imunologia , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Colo/metabolismo , Citocinas/antagonistas & inibidores , Citocinas/sangue , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Sulfato de Dextrana , Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/metabolismo , Masculino , Linfadenite Mesentérica/etiologia , Linfadenite Mesentérica/prevenção & controle , Camundongos Endogâmicos C57BL , Prebióticos/análise , Distribuição Aleatória , Proteína Amiloide A Sérica/análise , Proteína Amiloide A Sérica/antagonistas & inibidores , Solubilidade , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
3.
Respir Res ; 15: 46, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24735374

RESUMO

BACKGROUND: Asthma is estimated to affect as many as 300 million people worldwide and its incidence and prevalence are rapidly increasing throughout the world, especially in children and within developing countries. Recently, there has been a growing interest in the use of potentially beneficial bacteria for allergic diseases. This study is aimed at exploring the therapeutic effects of long-term treatment with two different beneficial bacterial strains (Bifidobacterium breve M-16 V and Lactobacillus rhamnosus NutRes1) and a glucocorticoid (budesonide), as a reference treatment, on inflammatory response in a murine model for chronic allergic asthma. METHODS: To mimic the chronic disease in asthmatic patients, we used the murine ovalbumin-induced asthma model combined with prolonged allergen exposure. Airway function; pulmonary airway inflammation; airway remodelling, mRNA expression of pattern recognition receptors, Th-specific cytokines and transcription factors in lung tissue; mast cell degranulation; in vitro T cell activation; and expression of Foxp3 in blood Th cells were examined. RESULTS: Lactobacillus rhamnosus reduced lung resistance to a similar extent as budesonide treatment in chronically asthmatic mice. Pulmonary airway inflammation, mast cell degranulation, T cell activation and airway remodelling were suppressed by all treatments. Beneficial bacteria and budesonide differentially modulated the expression of toll-like receptors (TLRs), nod-like receptors (NLRs), cytokines and T cell transcription factors. Bifidobacterium breve induced regulatory T cell responses in the airways by increasing Il10 and Foxp3 transcription in lung tissue as well as systemic by augmenting the mean fluorescence intensity of Foxp3 in blood CD4+ T cells. CONCLUSION: These findings show that Bifidobacterium breve M-16 V and Lactobacillus rhamnosus NutRes1 have strong anti-inflammatory properties that are comparable to budesonide and therefore may be beneficial in the treatment of chronic asthma.


Assuntos
Asma/tratamento farmacológico , Bifidobacterium , Budesonida/uso terapêutico , Modelos Animais de Doenças , Lacticaseibacillus rhamnosus , Pneumonia/tratamento farmacológico , Animais , Anti-Inflamatórios/uso terapêutico , Asma/microbiologia , Asma/patologia , Doença Crônica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/microbiologia , Pneumonia/patologia , Resultado do Tratamento
4.
PLoS One ; 9(2): e89121, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586537

RESUMO

Oral delivery of Gram positive bacteria, often derived from the genera Lactobacillus or Bifidobacterium, can modulate immune function. Although the exact mechanisms remain unclear, immunomodulatory effects may be elicited through the direct interaction of these bacteria with the intestinal epithelium or resident dendritic cell (DC) populations. We analyzed the immune activation properties of Lactobacilli and Bifidobacterium species and made the surprising observation that cellular responses in vitro were differentially influenced by the presence of serum, specifically the extracellular vesicle (EV) fraction. In contrast to the tested Lactobacilli species, tested Bifidobacterium species induce TLR2/6 activity which is inhibited by the presence of EVs. Using specific TLR ligands, EVs were found to enhance cellular TLR2/1 and TLR4 responses while TLR2/6 responses were suppressed. No effect could be observed on cellular TLR5 responses. We determined that EVs play a role in bacterial aggregation, suggesting that EVs interact with bacterial surfaces. EVs were found to slightly enhance DC phagocytosis of Bifidobacterium breve whereas phagocytosis of Lactobacillus rhamnosus was virtually absent upon serum EV depletion. DC uptake of a non-microbial substance (dextran) was not affected by the different serum fractions suggesting that EVs do not interfere with DC phagocytic capacity but rather modify the DC-microbe interaction. Depending on the microbe, combined effects of EVs on TLR activity and phagocytosis result in a differential proinflammatory DC cytokine release. Overall, these data suggest that EVs play a yet unrecognized role in host-microbe responses, not by interfering in recipient cellular responses but via attachment to, or scavenging of, microbe-associated molecular patterns. EVs can be found in any tissue or bodily fluid, therefore insights into EV-microbe interactions are important in understanding the mechanism of action of potential probiotics and gut immune homeostasis.


Assuntos
Bifidobacterium/fisiologia , Interações Hospedeiro-Patógeno , Fagocitose , Receptor 2 Toll-Like/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Bifidobacterium/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Ácido Láctico/biossíntese , Camundongos , Especificidade da Espécie , Receptor 6 Toll-Like/metabolismo , Vesículas Transportadoras/imunologia , Vesículas Transportadoras/microbiologia
5.
Biochim Biophys Acta ; 1842(4): 573-83, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24440361

RESUMO

Over the last decade, there has been a growing interest in the use of interventions that target the intestinal microbiota as a treatment approach for asthma. This study is aimed at exploring the therapeutic effects of long-term treatment with a combination of Bifidobacterium breve with non-digestible oligosaccharides on airway inflammation and remodeling. A murine ovalbumin-induced chronic asthma model was used. Pulmonary airway inflammation; mRNA expression of pattern recognition receptors, Th-specific cytokines and transcription factors in lung tissue; expression of Foxp3 in blood Th cells; in vitro T cell activation; mast cell degranulation; and airway remodeling were examined. The combination of B. breve with non-digestible oligosaccharides suppressed pulmonary airway inflammation; reduced T cell activation and mast cell degranulation; modulated expression of pattern recognition receptors, cytokines and transcription factors; and reduced airway remodeling. The treatment induced regulatory T cell responses, as shown by increased Il10 and Foxp3 transcription in lung tissue, and augmented Foxp3 protein expression in blood CD4+CD25+Foxp3+ T cells. This specific combination of beneficial bacteria with non-digestible oligosaccharides has strong anti-inflammatory properties, possibly via the induction of a regulatory T cell response, resulting in reduced airway remodeling and, therefore, may be beneficial in the treatment of chronic inflammation in allergic asthma.


Assuntos
Asma/tratamento farmacológico , Bifidobacterium , Oligossacarídeos/uso terapêutico , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Asma/imunologia , Doença Crônica , Citocinas/genética , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/análise , Masculino , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Receptores de Reconhecimento de Padrão/genética , Células Th2/imunologia
6.
J Leukoc Biol ; 94(5): 885-902, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23990624

RESUMO

TLRs play a major role in microbe-host interactions and innate immunity. Of the 10 functional TLRs described in humans, TLR2 is unique in its requirement to form heterodimers with TLR1 or TLR6 for the initiation of signaling and cellular activation. The ligand specificity of TLR2 heterodimers has been studied extensively, using specific bacterial and synthetic lipoproteins to gain insight into the structure-function relationship, the minimal active motifs, and the critical dependence on TLR1 or TLR6 for activation. Different from that for specific well-defined TLR2 agonists, recognition of more complex ligands like intact microbes or molecules from endogenous origin requires TLR2 to interact with additional coreceptors. A breadth of data has been published on ligand-induced interactions of TLR2 with additional pattern recognition receptors such as CD14, scavenger receptors, integrins, and a range of other receptors, all of them important factors in TLR2 function. This review summarizes the roles of TLR2 in vivo and in specific immune cell types and integrates this information with a detailed review of our current understanding of the roles of specific coreceptors and ligands in regulating TLR2 functions. Understanding how these processes affect intracellular signaling and drive functional immune responses will lead to a better understanding of host-microbe interactions and will aid in the design of new agents to target TLR2 function in health and disease.


Assuntos
Receptores Imunológicos/fisiologia , Receptor 2 Toll-Like/fisiologia , Animais , Humanos , Mediadores da Inflamação/fisiologia , Integrinas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Lectinas/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Lipopolissacarídeos/fisiologia , Microdomínios da Membrana/fisiologia , Polimorfismo de Nucleotídeo Único , Multimerização Proteica , Receptores CXCR4/fisiologia , Receptores de Reconhecimento de Padrão/fisiologia , Receptores Depuradores/fisiologia , Ácidos Teicoicos , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/química
7.
Mediators Inflamm ; 2013: 808470, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23781124

RESUMO

This study aimed at exploring innate and adaptive immunity in allergic asthma by investigation of mRNA expression of pattern recognition receptors, T-cell-specific cytokines, and transcription factors. Mouse models for mild and severe asthma, with similar pathological characteristics observed in humans, were used to study the involved inflammatory markers as a first step in the development of phenotype-directed treatment approaches. In the mild model, mice were sensitized to ovalbumin-Imject Alum and challenged with ovalbumin. In the severe model, mice were sensitized to trinitrophenyl-conjugated ovalbumin and challenged with trinitrophenyl-ovalbumin/IgE immune complex. Pulmonary airway inflammation and mRNA expression of Toll-like receptors (TLRs), NOD-like receptors (NLRs), T cell cytokines, and transcription factors in lung tissue were examined. Different mRNA expression profiles of TLRs, NLRs, T cell cytokines, and transcription factors were observed. In the mild model, Il10 showed the largest increase in expression, whereas in the severe model, it was Inf γ with the largest increase. Expression of Tbet was also significantly increased in the severe model. Inflammation and immunity are differentially regulated in mild and severe experimental asthma. This preclinical data may help in directing clinical research towards a better understanding and therapy in mild and severe asthmatic patients.


Assuntos
Asma/induzido quimicamente , Asma/imunologia , Animais , Modelos Animais de Doenças , Imunidade/efeitos dos fármacos , Imunidade/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/toxicidade , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Int Arch Allergy Immunol ; 159(1): 51-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22555211

RESUMO

BACKGROUND: Little is known about the contribution of the invariant natural killer T (iNKT) cells in the onset of food allergy. Using a mouse model for cow's milk allergy the function of iNKT cells was investigated. METHODS: Mice were sensitized orally with casein or whey proteins. One hour before the sensitizations the mice were injected intraperitoneally with α-galactosylceramide (αGalCer) or control. One week after the last sensitization acute allergic skin reactions were measured. Furthermore, in the liver, spleen and mesenteric lymph nodes (MLN) percentages of iNKT cells were analyzed and liver lymphocyte restimulation assays were performed. RESULTS: Whey- or casein-sensitized mice treated with αGalCer showed enhanced acute allergic skin reactions. The percentage of iNKT cells in the liver of sensitized mice was reduced compared to sham-sensitized mice. αGalCer treatment was found to deplete iNKT cells in the liver of sensitized as well as sham-sensitized mice, and these hepatocytes did not respond to ex vivo restimulation with αGalCer. αGalCer treatment did not reduce iNKT cell percentages in the spleen and MLN of sham-sensitized mice but abrogated the increase in iNKT cell percentage in the spleen upon whey sensitization, whereas it enhanced the iNKT cell percentage in the MLN of casein-sensitized mice. Due to the repeated application of αGalCer, livers were functionally depleted of iNKT cells. This resulted in an increased allergic effector response which was most pronounced in whey-sensitized mice and associated with enhanced whey-specific immunoglobulin levels. CONCLUSION: iNKT cells may suppress cow's milk allergic symptoms in mice and may differentially regulate oral sensitization for casein and whey.


Assuntos
Alérgenos/imunologia , Caseínas/imunologia , Hipersensibilidade a Leite/imunologia , Proteínas do Leite/imunologia , Células T Matadoras Naturais/imunologia , Animais , Células Cultivadas , Citocinas/imunologia , Feminino , Galactosilceramidas/farmacologia , Hepatócitos/imunologia , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos C3H , Hipersensibilidade a Leite/sangue , Proteínas do Soro do Leite
9.
J Nutr ; 141(5): 964-70, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21430245

RESUMO

Immune modulatory effects of EPA and DHA are well described. However, these fatty acids must be effectively incorporated into cell membrane phospholipids to modify cell function. To address the absence of human data regarding short-term incorporation, the present study investigated the incorporation of EPA and DHA into white blood cells (WBC) at different time points during 1 wk of supplementation with a medical food, which is high in protein and leucine and enriched with fish oil and specific oligosaccharides. Additionally, the effects on ex vivo immune function were determined. In a single-arm, open label study, 12 healthy men and women consumed 2 × 200 mL of medical food providing 2.4 g EPA, 1.2 g DHA, 39.7 g protein (including 4.4 g L-leucine), and 5.6 g oligosaccharides daily. Blood samples were taken at d 0 (baseline), 1, 2, 4, and 7. Within 1 d of nutritional intervention, the percentage of EPA in phospholipids of WBC increased from 0.5% at baseline to 1.3% (P < 0.001). After 1 wk, the percentage of EPA rose to 2.8% (P < 0.001). Additionally, the production of proinflammatory cytokines in LPS-stimulated whole blood cultures was significantly increased within 1 wk. Nutritional supplementation with a fish oil-enriched medical food significantly increased the percentage of EPA in phospholipids of WBC within 1 wk. Simultaneously, ex vivo immune responsiveness to LPS increased significantly. These results hold promise for novel applications such as fast-acting nutritional interventions in cancer patients, which should be investigated in future studies.


Assuntos
Proteínas Alimentares/administração & dosagem , Ácido Eicosapentaenoico/sangue , Óleos de Peixe/administração & dosagem , Alimentos Formulados , Imunomodulação , Leucócitos/metabolismo , Idoso , Transporte Biológico , Células Sanguíneas/imunologia , Células Sanguíneas/metabolismo , Células Cultivadas , Citocinas/sangue , Citocinas/metabolismo , Ácido Eicosapentaenoico/administração & dosagem , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Insaturados/administração & dosagem , Feminino , Alimentos Formulados/análise , Humanos , Leucócitos/imunologia , Masculino , Pessoa de Meia-Idade , Neoplasias/dietoterapia , Neoplasias/imunologia , Oligossacarídeos/administração & dosagem , Fosfolipídeos/química , Fatores de Tempo
10.
Int Rev Immunol ; 29(5): 485-513, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20839912

RESUMO

The intestinal immune system is severely affected by HIV and circulating microbial products from the intestinal tract that provide an ongoing source of systemic inflammation and concomitant viral replication. In addition, HIV-infected individuals can have a deregulated immune response that may hamper the anti-viral capacity of the host. Various probiotic organisms and prebiotic agents have been shown to enhance intestinal epithelial barrier functions, reduce inflammation, and support effective Th-1 responses. As these characteristics may benefit HIV patients, this review aims to provide a theoretical framework for the development of probiotic and prebiotic interventions specifically for this population.


Assuntos
Enteropatia por HIV/tratamento farmacológico , Infecções por HIV/tratamento farmacológico , HIV/fisiologia , Intestinos/microbiologia , Probióticos/uso terapêutico , HIV/patogenicidade , Enteropatia por HIV/imunologia , Enteropatia por HIV/microbiologia , Enteropatia por HIV/fisiopatologia , Infecções por HIV/imunologia , Infecções por HIV/microbiologia , Infecções por HIV/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade nas Mucosas , Inflamação , Intestinos/imunologia , Intestinos/virologia , Metagenoma/imunologia , Equilíbrio Th1-Th2 , Replicação Viral
11.
Int Immunopharmacol ; 10(5): 619-25, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20206301

RESUMO

The immune-modulatory effect of specific prebiotic oligosaccharides was shown in previous preclinical and clinical studies. To enhance the understanding of this effect, kinetic aspects of immune modulation and the correlation between microbiological and immunological parameters were investigated in a murine vaccination model. C57BL/6 mice were supplemented with short-chain galactooligosaccharides and long-chain fructooligosaccharides (ratio 9:1; Immunofortis()) in combination with pectin-derived acidic oligosaccharides. The timing of supplementation was varied around the primary (day 0) and secondary (day 21) vaccinations. Supplementation before the primary vaccination was necessary to increase delayed-type hypersensitivity responses (DTH) significantly at day 30. Supplementation after day 8 did not affect the DTH response at day 30, indicating that immune modulation occurred during the early phase. Therefore, correlation analysis of microbiological and immunological parameters was performed in a shortened experiment to focus on the early phase. At day 9 post-priming, the percentages of cecal lactobacilli were correlated to the DTH responses (p=0.01). Furthermore, the results suggested that yet unidentified factors may play a role. Additional analysis of intestinal Peyer's patch major lymphocyte populations did not show effects of supplementation. In conclusion, a specific oligosaccharide mixture was shown to exert its immune-modulatory effect during the early phase of a murine immune response. The results are consistent with a role of the microbiota and possibly other factors in oligosaccharide-induced immune modulation. Furthermore, the results demonstrate that it is critical to consider kinetic aspects of immune-modulatory and prebiotic effects in order to study their interaction in a meaningful way.


Assuntos
Hipersensibilidade Tardia/imunologia , Imunidade Celular/efeitos dos fármacos , Lactobacillus/imunologia , Oligossacarídeos/administração & dosagem , Pele/efeitos dos fármacos , Animais , Ceco/efeitos dos fármacos , Ceco/imunologia , Ceco/metabolismo , Ceco/microbiologia , Proliferação de Células/efeitos dos fármacos , DNA Bacteriano/análise , Feminino , Hipersensibilidade Tardia/patologia , Hipersensibilidade Tardia/fisiopatologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Lactobacillus/genética , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/química , Oligossacarídeos/imunologia , Nódulos Linfáticos Agregados/efeitos dos fármacos , Nódulos Linfáticos Agregados/patologia , Prebióticos , Pele/imunologia , Pele/patologia , Testes Cutâneos , Baço/efeitos dos fármacos , Baço/patologia , Vacinação
12.
Oncol Rep ; 22(6): 1403-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19885593

RESUMO

Cachexia and chronic inflammation are major challenges for cancer patients, leading to serious consequences. Accordingly, it is of high clinical relevance to identify early risk factors for optimal treatment, as these are currently not available. The present study demonstrates a strong decline in contact hypersensitivity, a parameter for cell-mediated immunity, in tumor-bearing cachectic mice. Interestingly, a significant reduction was already observed during the pre-cachectic state, reflecting an impaired immune function prior to weight loss. Extrapolating to the human setting, reduced immune competence of cancer patients could serve as an early marker for cancer cachexia, enabling an early supportive care strategy.


Assuntos
Caquexia/imunologia , Neoplasias/complicações , Neoplasias/imunologia , Animais , Biomarcadores , Caquexia/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Sistema Imunitário , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Monócitos/metabolismo , Neoplasias/patologia , Fatores de Risco , Fatores de Tempo
13.
Pediatr Allergy Immunol ; 18(4): 304-12, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17584310

RESUMO

Immunomodulatory effects of oligosaccharide preparations that resemble chemical and functional aspects of human milk oligosaccharides (HMOS) were studied for the development of new concepts in infant nutrition. A dose range of 1-5% (w/w) dietary pectin-derived acidic oligosaccharides (AOS) was tested in a murine influenza vaccination model. In addition, combinations of AOS and a 9:1 mixture of galacto-oligosaccharides and long-chain fructo-oligosaccharides (GOS/FOS) were tested at a fixed total dietary dose of 2% (w/w). It was found that AOS significantly enhanced vaccine-specific delayed-type hypersensitivity (DTH) responses in a dose-dependent manner. This was accompanied by a reduction in T-helper2 (Th2) cytokine production by splenocytes in vitro. Overall, this indicates that the systemic immune response to the vaccine was Th1-skewed by the dietary intervention. Combinations of GOS/FOS and AOS were more effective in enhancing DTH responses than either of the oligosaccharides alone, suggesting interaction effects between these agents. Similar to effects in infants, supplementation of the murine diets with GOS/FOS and combinations of GOS/FOS and AOS for 6-wk enhanced the proportion of fecal bifidobacteria and lactobacilli, but AOS alone did not. In conclusion, these data indicate that GOS/FOS and AOS enhance systemic Th1-dependent immune responses in a murine vaccination model. As Th1-responses are weak in early life in humans, this might suggest that application of these oligosaccharides in infant formulas will be beneficial for the development of the infant's immune system.


Assuntos
Suplementos Nutricionais , Vacinas contra Influenza/imunologia , Oligossacarídeos/imunologia , Células Th1/imunologia , Animais , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Oligossacarídeos/química , Células Th2/imunologia
14.
Int Immunopharmacol ; 6(8): 1277-86, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16782540

RESUMO

Analogous to reported immunomodulatory effects of probiotics, this study was performed to analyse the immunomodulatory properties of prebiotic oligosaccharides that share chemical characteristics with human milk oligosaccharides. A mixture containing galacto- and fructo-oligosaccharides (GOS/FOS; ratio 9:1) was tested at dietary doses between 1% and 10% (w/w of total diet) in an influenza vaccination model, using 10 C56BL/6JolaHsd mice per group. The modulation of vaccine specific delayed-type hypersensitivity (DTH) responses was studied as a marker of T-helper 1 (Th1) immunity, as well as other immune parameters. GOS/FOS enhanced DTH responses dose-dependently (optimum at 5% w/w of total diet; 41.4+/-14.1% increased compared to controls, p<0.05). No significant changes were detected on splenocyte proliferation or vaccine-specific antibody concentrations. Simultaneously, GOS/FOS dose-dependently increased the proportion of faecal bifidobacteria and lactobacilli (maximal effect at 10% w/w of total diet; 16.8+/-2.4% and 5.8+/-1.3% increased compared to controls respectively, p<0.01 for both parameters). In a comparative experiment, GOS/FOS and FOS/inulin (both at 2% w/w of total diet) induced similar significant effects on the gut microbiota. In contrast to GOS/FOS, FOS/inulin did not enhance DTH responses, indicating that an increase in the proportions of bifidobacteria and lactobacilli is not sufficient for an immunomodulatory effect in this model. The use of GOS/FOS in dietary products might provide an opportunity to stimulate the adaptive immune response in a Th1-direction and subsequently inhibit infections and Th2-related immune disorders in humans, for instance allergies. Clinical studies are being performed to confirm this.


Assuntos
Suplementos Nutricionais , Hipersensibilidade Tardia/imunologia , Oligossacarídeos/imunologia , Animais , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA Bacteriano/análise , DNA Bacteriano/genética , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Fezes/microbiologia , Feminino , Imunoglobulina G/sangue , Vacinas contra Influenza/imunologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/genética , Lactobacillus/isolamento & purificação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Oligossacarídeos/administração & dosagem , Probióticos/administração & dosagem , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Vacinação/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...