Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 8379, 2024 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-38600116

RESUMO

Macrophage responses to activation are fluid and dynamic in their ability to respond appropriately to challenges, a role integral to host defence. While bacteria can influence macrophage differentiation and polarization into pro-inflammatory and alternatively activated phenotypes through direct interactions, many questions surround indirect communication mechanisms mediated through secretomes derived from gut bacteria, such as lactobacilli. We examined effects of secretome-mediated conditioning on THP-1 human monocytes, focusing on the ability of the Lacticaseibacillus rhamnosus R0011 secretome (LrS) to drive macrophage differentiation and polarization and prime immune responses to subsequent challenge with lipopolysaccharide (LPS). Genome-wide transcriptional profiling revealed increased M2-associated gene transcription in response to LrS conditioning in THP-1 cells. Cytokine and chemokine profiling confirmed these results, indicating increased M2-associated chemokine and cytokine production (IL-1Ra, IL-10). These cells had increased cell-surface marker expression of CD11b, CD86, and CX3CR1, coupled with reduced expression of the M1 macrophage-associated marker CD64. Mitochondrial substrate utilization assays indicated diminished reliance on glycolytic substrates, coupled with increased utilization of citric acid cycle intermediates, characteristics of functional M2 activity. LPS challenge of LrS-conditioned THP-1s revealed heightened responsiveness, indicative of innate immune priming. Resting stage THP-1 macrophages co-conditioned with LrS and retinoic acid also displayed an immunoregulatory phenotype with expression of CD83, CD11c and CD103 and production of regulatory cytokines. Secretome-mediated conditioning of macrophages into an immunoregulatory phenotype is an uncharacterized and potentially important route through which lactic acid bacteria and the gut microbiota may train and shape innate immunity at the gut-mucosal interface.


Assuntos
Lacticaseibacillus rhamnosus , Monócitos , Humanos , Monócitos/metabolismo , Secretoma , Lipopolissacarídeos , Citocinas/metabolismo , Quimiocinas/metabolismo , Imunidade
2.
Front Microbiol ; 13: 980989, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36246229

RESUMO

Certain lactic acid bacteria (LAB) are associated with immune modulatory activities including down-regulation of pro-inflammatory gene transcription and expression. While host antigen-presenting cells (APCs) and intestinal epithelial cells (IEC) can interact directly with both pathogenic and commensal bacteria through innate immune pattern recognition receptors, recent evidence indicates indirect communication through secreted molecules is an important inter-domain communication mechanism. This communication route may be especially important in the context of IEC and APC interactions which shape host immune responses within the gut environment. We have previously shown that the Lacticaseibacillus rhamnosus R0011 secretome (LrS) dampens pro-inflammatory gene transcription and mediator production from Tumor Necrosis Factor-α and Salmonella enterica serovar Typhimurium secretome (STS)-challenged HT-29 IECs through the induction of negative regulators of innate immunity. However, many questions remain about interactions mediated through these bacterial-derived soluble components and the resulting host immune outcomes in the context of IEC and APC interactions. In the present study, we examined the ability of the LrS to down-regulate pro-inflammatory gene transcription and cytokine production from STS-challenged T84 human IEC and THP-1 human monocyte co-cultures. Cytokine and chemokine profiling revealed that apically delivered LrS induces apical secretion of macrophage inhibitory factor (MIF) and down-regulates STS-induced pro-inflammatory mediator secretion into the apical and basolateral chambers of the T84/THP-1 co-culture. Transcriptional profiling confirmed these results, as the LrS attenuated STS challenge-induced CXCL8 and NFκB1 expression in T84 IECs and THP-1 APCs. Interestingly, the LrS also reversed STS-induced damage to monolayer transepithelial resistance (TER) and permeability, results which were confirmed by ZO-1 gene expression and immunofluorescence visualization of ZO-1 expression in T84 IEC monolayers. The addition of a MIF-neutralizing antibody abrogated the ability of the LrS to reverse STS-induced damage to T84 IEC monolayer integrity, suggesting a novel role for MIF in maintaining IEC barrier function and integrity in response to soluble components derived from LAB. The results presented here provide mechanistic evidence for indirect communication mechanisms used by LAB to modulate immune responses to pathogen challenge, using in vitro approaches which allow for IEC and APC cell communication in a context which more closely mimics that which occurs in vivo.

3.
J Mech Behav Biomed Mater ; 120: 104583, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34062373

RESUMO

This paper focuses on utilizing the Fused Deposition Modeling (FDM) to manufacture Polycaprolactone/Nano-Hydroxyapatite/Chitin-Nano-Whisker nanocomposite scaffolds and their subsequent characterization for biomedical applications. FDM nanocomposite filaments were manufactured in multiple nanocomposite formulations of Polycaprolactone/Nano-Hydroxyapatite (nHA), Polycaprolactone/Chitin-Nano-Whisker (CNW), and Polycaprolactone/nHA/CNW using a green method. The FDM processing conditions were optimized using Taguchi orthogonal array method. The mechanical, biodegradation, and biocompatibility properties of the bone tissue scaffolds were assessed. A preosteoblast mouse bone cell line was used for cell proliferation and attachment assays. The results indicated that CNW content in the filaments slightly increases the mechanical properties of the 3D printed parts, and the nanocomposite with 3% CNW content exhibited significant improvement in the cell proliferation and attachment properties of the scaffolds. The nHA content considerably improved the mechanical properties of the scaffolds. The nHA and CNW nanofillers increased the biodegradation rate of PCL. In general, considering all types of responses, a green manufactured nanocomposite of PCL/nHA/CNW can significantly increase the biological and mechanical properties of the 3D printed products for bone tissue scaffolds.


Assuntos
Nanocompostos , Tecidos Suporte , Animais , Quitina , Durapatita , Camundongos , Poliésteres , Engenharia Tecidual
4.
Curr Res Food Sci ; 3: 51-58, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32914120

RESUMO

Fermented dairy products have become attractive functional foods for the delivery of probiotics and their biologically active metabolites. The aim of this study was to examine the immunomodulatory activity of milk fermented with the probiotic lactic acid bacterium Lactobacillus rhamnosus R0011 (LrF) on macrophages challenged with lipopolysaccharide (LPS), a potent pro-inflammatory stimulus. To this end, human THP-1 or U937 monocytes were differentiated into resting macrophages then stimulated with LPS and co-incubated with the LrF or with milk controls. Levels of pro-inflammatory and immunoregulatory cytokines were determined by enzyme-linked immunosorbent assays. Culturing of LPS-stimulated U937 macrophages with either the whole or filtered LrF resulted in an increase in Interleukin (IL)-1Ra production relative to the negative control. THP-1 macrophages cultured with the LrF demonstrated an increase in LPS-induced IL-10 and IL-1ß production, while production of LPS-induced IL-6, sCD54, IL-8, IL-1ß, TNF-α, IL-12p70 and Transforming Growth Factor-ß (TGF-ß) was unaffected. Further, the LrF induced the expression of DC-SIGN and CD206, markers of immunoregulatory M2 macrophage polarization, in LPS-challenged THP-1 macrophages. Taken together, milk fermented with L. rhamnosus R0011 increased regulatory cytokine production from LPS-challenged U937 and THP-1 macrophages, while simultaneously up-regulating the production of IL-1ß and expression of DC-SIGN and CD206, a profile characteristic of polarization into the immunoregulatory M2 macrophage phenotype.

5.
J Immunol ; 204(9): 2523-2534, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238458

RESUMO

Recent evidence suggests that lactic acid bacteria communicate with host cells via secretome components to influence immune responses but less is known about gut-pathogen secretomes, impact of lactic acid bacteria secretomes on host-pathogen interactions, and the mechanisms underlying these interactions. Genome-wide microarrays and cytokine profiling were used to interrogate the impact of the Lactobacillus rhamnosus R0011 secretome (LrS) on TNF-α and Salmonella enterica subsp. enterica serovar Typhimurium secretome (STS)-induced outcomes in human intestinal epithelial cells. The LrS attenuated both TNF-α- and STS-induced gene expression involved in NF-κB and MAPK activation, as well as expression of genes involved in other immune-related signaling pathways. Specifically, the LrS induced the expression of dual specificity phosphatase 1 (DUSP1), activating transcription factor 3 (ATF3), and tribbles pseudokinase 3 (TRIB3), negative regulators of innate immune signaling, in HT-29 intestinal epithelial cells challenged with TNF-α or STS. TNF-α- and STS-induced acetylation of H3 and H4 histones was attenuated by the LrS, as was the production of TNF-α- and STS-induced proinflammatory cytokines and chemokines. Interestingly, the LrS induced production of macrophage migration inhibitory factor (MIF), a cytokine involved in host-microbe interactions at the gut interface. We propose that the LrS attenuates proinflammatory mediator expression through increased transcription of negative regulators of innate immune activity and changes in global H3 and H4 histone acetylation. To our knowledge, these findings provide novel insights into the complex multifaceted mechanisms of action behind secretome-mediated interdomain communication at the gut-mucosal interface.


Assuntos
Células Epiteliais/imunologia , Inflamação/imunologia , Intestinos/imunologia , Lacticaseibacillus rhamnosus/imunologia , Salmonelose Animal/imunologia , Salmonella typhimurium/imunologia , Fator de Necrose Tumoral alfa/imunologia , Acetilação , Animais , Linhagem Celular Tumoral , Citocinas/imunologia , Células Epiteliais/microbiologia , Expressão Gênica/imunologia , Células HT29 , Histonas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/imunologia , Inflamação/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Salmonelose Animal/microbiologia , Sorogrupo , Transdução de Sinais/fisiologia , Transcrição Gênica/imunologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-31636910

RESUMO

BACKGROUND: Evidence suggests that disrupting prolonged bouts of sitting with short bouts of physical activity can significantly reduce blood glucose and improve insulin sensitivity; however, limited research is available on the impact of such disruptions on inflammation and swelling. The purpose of this study was to determine whether short bouts of exercise performed each hour during a 4 h sitting session were able to negate the effects of prolonged sitting (PS) on several cardiometabolic outcomes. METHODS: Eligible participants (n = 10) attended two laboratory sessions: PS (uninterrupted sitting for 4 h) and disrupted sitting (DS; 4 h sitting session disrupted by 3 min of exercise each hour (60-s warm-up at 50 W, 5 s of unloaded cycling, 20-s sprint at 5% body weight, and 95-s cool-down at 50 W)). The exercise bouts were performed at minute 60, 120, and 180. Blood and saliva samples, and measures of heart rate and blood pressure were assessed before (T1) and after (T2) each session; leg swell was measured continuously. RESULTS: Concentrations of salivary IL-8 increased during PS (T1: 0.19 ± 0.32; T2: 0.50 ± 1.00 pg/µg of protein) but decreased during DS (T1: 0.41 ± 0.23; T2: 0.22 ± 0.11 pg/µg of protein, d: 0.51, p = 0.002). Leg swell increased and plateaued in PS, but was attenuated during DS. CONCLUSION: It appears that short bouts of exercise significantly reduce swelling in the lower leg and IL-8 levels in the saliva, indicating that even among healthy, active, young adults, disrupting prolonged sitting can significantly reduce swelling and systemic inflammation.

7.
Front Immunol ; 9: 2639, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524427

RESUMO

Host intestinal epithelial cells (IEC) present at the gastrointestinal interface are exposed to pathogenic and non-pathogenic bacteria and their products. Certain probiotic lactic acid bacteria (LAB) have been associated with a range of host-immune modulatory activities including down-regulation of pro-inflammatory gene expression and cytokine production by IEC, with growing evidence suggesting that these bacteria secrete bioactive molecules with immunomodulatory activity. The aim of this study was to determine whether two lactobacilli with immunomodulatory activity [Lactobacillus rhamnosus R0011 (Lr) and Lactobacillus helveticus R0389 (Lh)], produce soluble mediators able to influence IEC responses to Pattern Recognition Receptor (PRR) ligands and pro-inflammatory cytokines [Tumor Necrosis Factor α (TNFα), Interleukin-1ß (IL-1ß)], signals inducing IEC chemokine production during infection. To this end, the effects of cell-free supernatants (CFS) from Lr and Lh on IEC production of the pro-inflammatory chemokines interleukin (IL)-8 and cytokine-induced neutrophil chemoattractant 1 (CINC-1) induced by a range of host- or pathogen-derived pro-inflammatory stimuli were determined, and the impact on human HT-29 IEC and a primary IEC line (rat IEC-6) was compared. The Lr-CFS and Lh-CFS did not significantly modulate basal IL-8 production from HT-29 IECs or CINC-1 production from IEC-6 cells. However, both Lr-CFS and Lh-CFS significantly down-regulated IL-8 production from HT-29 IECs challenged with varied PRR ligands. Lr-CFS and Lh-CFS had differential effects on PRR-induced CINC-1 production by rat IEC-6 IECs, with no significant down-regulation of CINC-1 observed from IEC-6 IECs cultured with Lh-CFS. Further analysis of the Lr-CFS revealed down-regulation of IL-8 production induced by the pro-inflammatory cytokines IL-1ß and TNFα Preliminary characterization of the bioactive constituent(s) of the Lr-CFS indicates that it is resistant to treatment with DNase, RNase, and an acidic protease, but is sensitive to alterations in pH. Taken together, these results indicate that these lactobacilli secrete bioactive molecules of low molecular weight that may modulate host innate immune activity through interactions with IEC.


Assuntos
Quimiocinas/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Lacticaseibacillus rhamnosus/imunologia , Lactobacillus helveticus/imunologia , Animais , Linhagem Celular Tumoral , Humanos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...