Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
1.
Biochem Biophys Res Commun ; 731: 150279, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39018972

RESUMEN

This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARß, PPARγ, C/EBPα, C/EBPß, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.

2.
J Agric Food Chem ; 72(32): 17924-17937, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38965062

RESUMEN

Turmeric, derived from Curcuma longa, and Lactobacillus paracasei, a lactic acid bacteria, have been studied for their potential antiobesity effects. To date, the antiobesity effects of turmeric fermented with L. paracasei have not been sufficiently investigated. This study was conducted via oral administration of 5% L. paracasei-fermented (FT) and unfermented turmeric (UT) in diet over 16 weeks using high-fat diet (HFD)-induced obese C57BL/6J mice. Results showed that the curcuminoid content of turmeric decreased following fermentation. Furthermore, FT significantly suppressed weight gain and liver and visceral adipose tissue weight and reduced plasma metabolic parameters in both the UT and FT experimental groups. The effects of FT were more noticeable than those of the unfermented form. Moreover, FT downregulated the expression of adipogenesis, lipogenesis, and inflammatory-related protein, but upregulated liver ß-oxidation protein SIRT 1, PPARα, and PGC-1α in perigonadal adipose tissue. Additionally, FT ameliorated insulin resistance by activating insulin receptor pathway protein expressions in visceral adipose tissues. FT also modulated gut microbiota composition, particularly in two beneficial bacteria, Akkermansia muciniphila and Desulfovibrio, as well as two short-chain fatty acid-producing bacteria: Muribaculum intestinale and Deltaproteobacteria. Our findings indicate that the modulation effect of FT may be an important pathway for its antiobesity mechanisms.


Asunto(s)
Curcuma , Dieta Alta en Grasa , Fermentación , Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Ratones Endogámicos C57BL , Obesidad , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Obesidad/microbiología , Ratones , Curcuma/química , Curcuma/metabolismo , Masculino , Lacticaseibacillus paracasei/metabolismo , Humanos , Fármacos Antiobesidad/administración & dosificación , Adipogénesis/efectos de los fármacos , Hígado/metabolismo , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , PPAR alfa/metabolismo , PPAR alfa/genética , Resistencia a la Insulina , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Bacterias/efectos de los fármacos
3.
Meat Sci ; 216: 109578, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38917677

RESUMEN

Novel shelf-stable and high-protein meat products that are affordable, convenient, and healthy are hot topic in current food innovation trends. To offer technological databases for developing new functional lamb meat products, this study aimed to evaluate the technological and sensory aspects of dry-cured lamb meat snacks incorporated with the probiotic culture Lactobacillus paracasei and the prebiotic lactulose. Four formulations were analyzed: control (without prebiotic or probiotic); PREB (with 2% lactulose); PROB (with 107 CFU/g of L. paracasei); and SYMB (with 2% lactulose and 107 CFU/g of L. paracasei). Fitted curves revealed that weight-loss behavior during snack ripening was not affected (P > 0.05) by treatments. Snack moisture, water activity, pH, titratable acidity, lipid oxidation, and residual nitrite were affected (P < 0.05) only by ripening time. The target probiotic strain stood out against competitive flora and was detected at 107 CFU/g in the snack-supplemented formulations (PROB and SYMB). In snacks supplemented with prebiotics (PREB and SYMB), the lactulose content was maintained at 2.17%. Significant differences were not observed in the chemical composition, texture profiles, and CIE color indices between the proposed functional snacks and the control. In addition to texture, flavor, and overall impression evaluation, only color attributes were positively impacted (P < 0.05) in the acceptance and multiple comparison tests against the control. The proposed formulation and bench process parameters produced potential nutritionally and sensory-appreciated, microbiologically stable, and safe (multi-hurdle perspective) functional high-protein restructured lamb snacks.


Asunto(s)
Productos de la Carne , Prebióticos , Probióticos , Oveja Doméstica , Bocadillos , Gusto , Animales , Productos de la Carne/microbiología , Productos de la Carne/análisis , Humanos , Lactobacillus , Lactulosa , Manipulación de Alimentos/métodos , Carne Roja/microbiología , Carne Roja/análisis , Color , Masculino , Femenino
4.
Oncologist ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38767987

RESUMEN

BACKGROUND: Abemaciclib-induced diarrhea is a relevant concern in clinical practice. Postbiotics have emerged as a promising option for managing it. MATERIALS AND METHODS: We conducted a retrospective-prospective, 2-group, observational study to assess the impact of the postbiotic PostbiotiX-Restore, derived by Lactobacillus paracasei CNCM I-5220, on abemaciclib-induced diarrhea in patients with hormone receptor-positive HER2-negative breast cancer. The prospective population (Postbio group) received postbiotic during the first cycle of abemaciclib, while the retrospective one received standard care (Standard group). Diarrhea grading was defined according to the National Cancer Institute's Common Terminology Criteria for Adverse Events. RESULTS: During the first cycle, diarrhea occurred in 78.9% of patients in the Standard cohort and 97.1% in the Postbio one, with most cases being G1-G2. Severe (G3) diarrhea was significantly less frequent in the Postbio group (0%) compared to the Standard one (7.9%; P = .029). Over the entire study period, while the grading difference was not statistically significant, G3 events were less frequent in the Postbio population (5.9%) than the Standard one (15.4%). Moreover, Postbio patients required fewer dose reductions due to diarrhea compared to the Standard group (P = .002). Notably, in the Postbio population, G1 and G2 events had short median durations (3 and 1 days, respectively) and, for the 2 patients experiencing G3 events during the second abemaciclib cycle (off postbiotic), diarrhea lasted only 1 day. CONCLUSIONS: Our study demonstrates the effect of PostbiotiX-Restore in mitigating abemaciclib-induced diarrhea, resulting in reduced severity, fewer dose reductions, and shorter duration. Further exploration and validation in larger cohorts are needed.

5.
Artif Cells Nanomed Biotechnol ; 52(1): 278-290, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38733373

RESUMEN

Type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD), obesity (OB) and hypertension (HT) are categorized as metabolic disorders (MDs), which develop independently without distinct borders. Herein, we examined the gut microbiota (GM) and Saururus chinensis (SC) to confirm their therapeutic effects via integrated pharmacology. The overlapping targets from the four diseases were determined to be key protein coding genes. The protein-protein interaction (PPI) networks, and the SC, GM, signalling pathway, target and metabolite (SGSTM) networks were analysed via RPackage. Additionally, molecular docking tests (MDTs) and density functional theory (DFT) analysis were conducted to determine the affinity and stability of the conformer(s). TNF was the main target in the PPI analysis, and equol derived from Lactobacillus paracasei JS1 was the most effective agent for the formation of the TNF complex. The SC agonism (PPAR signalling pathway), and antagonism (neurotrophin signalling pathway) by SC were identified as agonistic bioactives (aromadendrane, stigmasta-5,22-dien-3-ol, 3,6,6-trimethyl-3,4,5,7,8,9-hexahydro-1H-2-benzoxepine, 4α-5α-epoxycholestane and kinic acid), and antagonistic bioactives (STK734327 and piclamilast), respectively, via MDT. Finally, STK734327-MAPK1 was the most favourable conformer according to DFT. Overall, the seven bioactives from SC and equol that can be produced by Lactobacillus paracasei JS1 can exert synergistic effects on these four diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Hipertensión , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Saururaceae , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/microbiología , Obesidad/metabolismo , Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipertensión/microbiología , Hipertensión/metabolismo , Hipertensión/tratamiento farmacológico , Animales , Saururaceae/química , Saururaceae/metabolismo , Simulación del Acoplamiento Molecular , Humanos , Mapas de Interacción de Proteínas
6.
Eur J Nutr ; 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38733401

RESUMEN

PURPOSE: Ulcerative colitis (UC) is a serious health problem with increasing morbidity and prevalence worldwide. The pathogenesis of UC is complex, currently believed to be influenced by genetic factors, dysregulation of the host immune system, imbalance in the intestinal microbiota, and environmental factors. Currently, UC is typically managed using aminosalicylates, immunosuppressants, and biologics as adjunctive therapies, with the risk of relapse and development of drug resistance upon discontinuation. Therefore, further research into the pathogenesis of UC and exploration of potential treatment strategies are necessary to improve the quality of life for affected patients. According to previous studies, Lactobacillus paracasei Jlus66 (Jlus66) reduced inflammation and may help prevent or treat UC. METHODS: We used dextran sulfate sodium (DSS) to induce a mouse model of UC to assess the effect of Jlus66 on the progression of colitis. During the experiment, we monitored mouse body weight, food and water consumption, as well as rectal bleeding. Hematoxylin-eosin staining was performed to assess intestinal pathological damage. Protein imprinting and immunohistochemical methods were used to evaluate the protein levels of nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase (MAPK), and tight junction (TJ) proteins in intestinal tissues. Fecal microbiota was analyzed based on partial 16S rRNA gene sequencing. RESULTS: Jlus66 supplementation reduced the degree of colon tissue damage, such as colon shortening, fecal occult blood, colon epithelial damage, and weight loss. Supplementation with Jlus66 reduced DSS-induced upregulation of cytokine levels such as TNF-α, IL-1ß, and IL-6 (p < 0.05). The NF-κB pathway and MAPK pathway were inhibited, and the expression of TJ proteins (ZO-1, Occludin, and Claudin-3) was upregulated. 16S rRNA sequencing of mouse cecal contents showed that Jlus66 effectively regulated the structure of the intestinal biota. CONCLUSION: In conclusion, these data indicate that Jlus66 can alter the intestinal biota and slow the progression of UC, providing new insights into potential therapeutic strategies for UC.

7.
Artículo en Inglés | MEDLINE | ID: mdl-38658186

RESUMEN

Lactobacillus paracasei IMC502® is a commercially successful probiotic strain. However, there are no reports that investigate growth medium composition in relation to improved biomass production for this strain. The major outcome of the present study is the design and optimization of a growth medium based on vegan components to be used in the cultivation of Lactobacillus paracasei IMC502®, by using Design of Experiments. Besides comparing different carbon sources, the use of plant-based peptones as nitrogen sources was considered. In particular, the use of guar peptone as the main nitrogen source, in the optimization of fermentation media for the production of probiotics, could replace other plant peptones (e.g. potato, rice, wheat, and soy) which are part of the human diet, thereby avoiding an increase in product and process prices. A model with R2 and adjusted R2 values higher than 95% was obtained. Model accuracy was equal to 94.11%. The vegan-optimized culture medium described in this study increased biomass production by about 65% compared to growth on De Man-Rogosa-Sharpe (MRS) medium. Moreover, this approach showed that most of the salts and trace elements generally present in MRS are not affecting biomass production, thus a simplified medium preparation can be proposed with higher probiotic biomass yield and titer. The possibility to obtain viable lactic acid bacteria at high density from vegetable derived nutrients will be of great interest to specific consumer communities, opening the way to follow this approach with other probiotics of impact for human health.


Asunto(s)
Medios de Cultivo , Fermentación , Lacticaseibacillus paracasei , Probióticos , Medios de Cultivo/química , Probióticos/metabolismo , Lacticaseibacillus paracasei/metabolismo , Lacticaseibacillus paracasei/crecimiento & desarrollo , Biomasa , Nitrógeno/metabolismo , Peptonas/metabolismo , Carbono/metabolismo
8.
Cell Biochem Funct ; 42(2): e3975, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38475877

RESUMEN

Different organic compounds can have varying degrees of impact on the activity of Lactobacillus paracasei. The study focused on the impact and action mechanism of different organic selenium products on the bioactivity of two strains of L. paracasei. The growth, antioxidant activity, extracellular polysaccharide secretion, quorum sensing (QS), and biofilm formation of the strains before and after the addition of organic selenium crude products and three organic selenium standard were evaluated. The results showed that the addition of crude organic selenium promoted the various activities of the strain. l-selenocysteine had the strongest regulatory effect, with maximum GIM1.80 biofilm formation when it reached a critical concentration of 0.4 µg/mL; l-selenomethionine resulted in the highest activity of the signal molecule Auto inducer-2 of GDMCC1.155, when it reached a critical concentration of 0.4 µg/mL. The results of scanning electron microscopy demonstrated that the addition of organic selenium effectively improved the morphological structure of the two bacterial cells. Molecular docking revealed that the mechanism by which organic selenium regulates QS in Lactobacillus was achieved by binding two crucial receptor proteins (histidine protein kinase HKP and periplasmic binding protein LuxP) from specific sites. Furthermore, organic selenium products have a beneficial regulatory effect on the biological activity of L. paracasei. Overall, these findings provide a new alternative (organic selenium) for regulating the viability and beneficial activity of L. paracasei.


Asunto(s)
Lacticaseibacillus paracasei , Selenio , Percepción de Quorum , Antioxidantes/farmacología , Selenio/farmacología , Simulación del Acoplamiento Molecular , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Biopelículas
9.
J Dairy Sci ; 107(8): 5280-5300, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38460876

RESUMEN

Gut microbiota imbalance could lead to various diseases, making it important to optimize the structure of the gut flora in adults. Lactobacillus paracasei ZFM54 is a bacteriocin- and folic acid-producing Lactobacillus strain. Herein, L. paracasei ZFM54 was used as the potentially probiotic bacterium to ferment milk together with a yogurt starter. We optimized the fermentation conditions, and the obtained yogurts were then subjected to volatile and nonvolatile metabolome analysis, showing that L. paracasei ZFM54 can not only improve the acidity, water holding capacity and live lactic acid bacteria counts, but also improve many volatile acid contents and increase some beneficial nonvolatile metabolites, such as N-ethyl glycine and l-lysine, endowing the yogurt with more flavor and better function. The regulatory effects of the co-fermented yogurt on the intestinal microecology of volunteers were investigated by 16S rRNA sequencing and short-chain fatty acid (SCFA) analysis after consuming the yogurt for a 2-wk period, showing a better effect to increase the relative abundance of beneficial bacteria such as Ruminococcus and Alistipes, decrease harmful bacteria (Escherichia-Shigella and Enterobacter), and enhance the production of SCFA (acetate, propionate, and butyric acid) compared with the control yogurt. We found that L. paracasei ZFM54 can significantly improve the health benefits of yogurt, laying the foundation for its commercial application in improving gut microbiota.


Asunto(s)
Fermentación , Microbioma Gastrointestinal , Yogur , Yogur/microbiología , Humanos , Adulto , Probióticos , Lacticaseibacillus paracasei/metabolismo , Ácidos Grasos Volátiles/metabolismo , Lactobacillus/metabolismo
10.
Toxins (Basel) ; 16(2)2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38393172

RESUMEN

Probiotics and their bacteriocins have increasingly attracted interest for their use as safe food preservatives. This study aimed to produce soft white cheese fortified with Lacticaseibacillus MG847589 (Lb. paracasei MG847589) and/or its bacteriocin; cheese with Lacticaseibacillus (CP), cheese with bacteriocin (CB), and cheese with both Lacticaseibacillus and bacteriocin (CPB) were compared to control cheese (CS) to evaluate their biopreservative and anti-mycotoxigenic potentials for prolonged shelf life and safe food applications. The effects of these fortifications on physiochemical, microbial, texture, microstructure, and sensory properties were studied. Fortification with Lacticaseibacillus (CP) increased acidity (0.61%) and microbial counts, which may make the microstructure porous, while CPB showed intact microstructure. The CPB showed the highest hardness value (3988.03 g), while the lowest was observed with CB (2525.73 g). Consequently, the sensory assessment reflected the panelists' preference for CPB, which gained higher scores than the control (CS). Fortification with Lb. paracasei MG847589 and bacteriocin (CPB) showed inhibition effects against S. aureus from 6.52 log10 CFU/g at time zero to 2.10 log10 CFU/g at the end of storage, A. parasiticus (from 5.06 to 3.03 log10 CFU/g), and P. chrysogenum counts (from 5.11 to 2.86 log10 CFU/g). Additionally, CPB showed an anti-mycotoxigenic effect against aflatoxins AFB1 and AFM1, causing them to be decreased (69.63 ± 0.44% and 71.38 ± 0.75%, respectively). These potentials can extend shelf life and pave the way for more suggested food applications of safe food production by fortification with both Lb. paracasei MG847589 and its bacteriocin as biopreservatives and anti-mycotoxigenic.


Asunto(s)
Bacteriocinas , Queso , Lacticaseibacillus paracasei , Lactobacillus , Bacteriocinas/farmacología , Staphylococcus aureus , Microbiología de Alimentos
11.
Bioact Mater ; 35: 31-44, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38304916

RESUMEN

Skin microbiota plays an important role in wound healing, but skin injuries are highly susceptible to wound infections, leading to disruption of the skin microbiota. However, conventional antibacterial hydrogels eliminate both probiotics and pathogenic bacteria, disrupting the balance of the skin microbiota. Therefore, it is important to develop a wound dressing that can fend off foreign pathogenic bacteria while preserving skin microbiota stability. Inspired by live bacteria therapy, we designed a probiotic hydrogel (HAEPS@L.sei gel) with high viability for promoting wound healing. Lactobacillus paracasei TYM202 encapsulated in the hydrogel has the activity of promoting wound healing, and the hydrogel matrix EPS-M76 has the prebiotic activity that promotes the proliferation and metabolism of Lactobacillus paracasei TYM202. During the wound healing process, HAEPS@L.sei gel releases lactic acid and acetic acid to resist the growth of pathogenic bacteria while maintaining Firmicutes and Proteobacteria balance at the phylum level, thus preserving skin microbiota stability. Our results showed that live probiotic hydrogels reduce the incidence of inflammation during wound healing while promoting angiogenesis and increasing collagen deposition. This study provides new ideas for developing wound dressings predicated on live bacterial hydrogels.

12.
Metabolites ; 14(1)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38276307

RESUMEN

Human milk is the gold standard for infant nutrition, but when it is not available or insufficient to satisfy the needs of the infant, formula milk is proposed as an effective substitute. A prospective observational cohort study was conducted on late preterm infants fed with breast and two different formula milks. On this basis, they were divided into three groups: group FMPB (fed with formula + postbiotic), group FM (fed with standard formula), and group BM (breastfed). Stool samples for a metabolomic study were collected at T0 (5-7 days after birth), T1 (30 days of life), and T2 (90 days of life), giving rise to 74 samples analyzed via liquid chromatography coupled with high-resolution mass spectrometry. The T0, T1, and T2 LC-MS raw data were processed for Partial Least Square Discriminant Analysis (PLS-DA), followed by a statistical analysis. This preliminary study highlighted a good overlapping between the fecal metabolome of breast and substitute feeding systems, confirming the efficacy of the formula preparations as breast milk substitutes. Moreover, several similarities were also detected between the FMPB and BM metabolome, highlighting that the addition of a postbiotic to standard formula milk could be more effective and considered a better alternative to breast milk.

13.
Nutrients ; 16(2)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38257160

RESUMEN

Glycolipid metabolic disorders (GLMD) refer to a series of metabolic disorders caused by abnormal processes of glucose and lipid synthesis, decomposition, and absorption in the body, leading to glucose and lipid excess, insulin resistance, and obesity. Probiotic intervention is a new strategy to alleviate metabolic syndrome. Lactobacillus paracasei JY062 (L. paracasei JY062) was separated from the Tibet-fermented dairy products. The results demonstrated a strong ability to relieve blood glucose disorders, blood lipid disorders, and tissue damage. The LPH group had the best effect, significantly decreasing the total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), leptin, insulin, and free fatty acid (FFA) concentrations and increasing the high-density lipoprotein cholesterol, adiponectin, and GLP-1 level compared to HFD-group mice. L. paracasei JY062 could activate the APN-AMPK pathway, increased AdipoQ, AMPK GLUT-4, and PGC-1α mRNA expression and decreased SREBP-1c, ACC, and FAS mRNA expression. L. paracasei JY062 intervention decreased the relative abundance of harmful bacteria, increased the relative abundance of beneficial bacteria, and restored the imbalance of gut microbiota homeostasis caused by a high-glucose-fat diet. L. paracasei JY062 alleviated glucolipid metabolism disorders via the adipoinsular axis and gut microbiota. This study provided a theoretical basis for probiotics to ameliorate glucolipid metabolism disorders by regulating the adipoinsular axis.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Enfermedades Metabólicas , Animales , Ratones , Proteínas Quinasas Activadas por AMP , HDL-Colesterol , Glucosa , ARN Mensajero
14.
Arch Pharm (Weinheim) ; 357(1): e2300422, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37861276

RESUMEN

Pineapple has been recognized for its potential to enhance health and well-being. This study aimed to gain molecular insights into the anti-inflammatory properties of fermented pineapple juice using multimodal computational studies. In this study, pineapple juice was fermented using Lactobacillus paracasei, and the solution underwent liquid chromatography-mass spectrometry analysis. Network pharmacology was applied to investigate compound interactions and targets. In silico methods assessed compound bioactivities. Protein-protein interactions, network topology, and enrichment analysis identified key compounds. Molecular docking explored compound-receptor interactions in inflammation regulation. Molecular dynamics simulations were conducted to confirm the stability of interactions between the identified crucial compounds and their respective receptors. The study revealed several compounds including short-chain fatty acids, peptides, dihydroxyeicosatrienoic acids, and glycerides that exhibited promising anti-inflammatory properties. Leucyl-leucyl-norleucine and Leu-Leu-Tyr exhibited robust and stable interactions with mitogen-activated protein kinase 14 and IκB kinase ß, respectively, indicating their potential as promising therapeutic agents for inflammation modulation. This proposition is grounded in the pivotal involvement of these two proteins in inflammatory signaling pathways. These findings provide valuable insights into the anti-inflammatory potential of these compounds, serving as a foundation for further experimental validation and exploration. Future studies can build upon these results to advance the development of these compounds as effective anti-inflammatory agents.


Asunto(s)
Ananas , Ananas/química , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Antiinflamatorios/farmacología , Inflamación
15.
Allergol Int ; 73(2): 302-312, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37953104

RESUMEN

BACKGROUND: Lactobacillus paracasei has been known to reduce airway resistance and inflammation in asthma. However, the therapeutic effect of its extracellular vesicles (EVs) in patients with asthma remains unclear. METHODS: To validate the clinical relevance of L. paracasei-derived EVs (LpEV) in asthma, the composition of gut microbial EVs was verified by metagenomics in LPS-induced C57BL/6 mice. The components of proteins and metabolites in LpEV were identified by peptide mass fingerprinting and metabolomic analysis. The serum levels of specific IgG1 or IgG4 antibodies to LpEV were compared by ELISA between patients with eosinophilic asthma (EA, n = 10) and those with neutrophilic asthma (NA, n = 10) as well as with healthy controls (HCs, n = 10). Finally, therapeutic effects of LpEV and their metabolites in asthma were validated in vivo/in vitro. RESULTS: Significantly lower proportions of EVs derived from Lactobacillus at the genus level were noted in mice with NA than in control mice. Moreover, the serum levels of LpEV-specific IgG4, but not IgG1, were lower in patients with NA than in those with EA or in HCs and positively correlated with FEV1 (%) values. In addition, oral administration of LpEV reduced airway resistance and inflammation in mice with NA. Finally, LpEV and their 3 metabolites (dodecanoic acid, palmitoleic acid, and D-(-)-tagatose) significantly inhibited JNK phosphorylation/IL-8 production in airway epithelium in vitro. CONCLUSIONS: These findings suggest that LpEV may have a therapeutic potential targeting NA by suppressing the JNK pathway and proinflammatory cytokine production in airway epithelium.


Asunto(s)
Asma , Vesículas Extracelulares , Lacticaseibacillus paracasei , Animales , Humanos , Ratones , Asma/terapia , Modelos Animales de Enfermedad , Epitelio , Inmunoglobulina G , Inflamación , Pulmón , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL
16.
J Sci Food Agric ; 104(2): 664-674, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-37653286

RESUMEN

BACKGROUND: A large number of people worldwide suffer from gluten-induced food allergy. As investigated in our previous research, Lactobacillus paracasei AH2 isolated from traditionally homemade sourdough in Anhui province of China showed the potential to reduce the immune reactivity of wheat protein by in vitro evaluation. However, whether L. paracasei AH2 has a role in alleviating wheat allergy in an in vivo model and its underlying mechanisms have not been elucidated. RESULTS: In this study, the alleviative effects of L. paracasei AH2 on gluten-induced allergic response were evaluated. Compared with a gluten-allergic mouse, L. paracasei AH2 suppressed anaphylaxis symptoms, gluten-specific immunoglobulin E, histamine and interleukin-4. Moreover, L. paracasei AH2 attenuated splenomegaly and induced Th1 or Treg cell differentiation to modulate the Th1/Th2 immune balance toward Th1 polarization. Short-chain fatty acid (SCFA) levels were enhanced after L. paracasei AH2 supplementation, contributing to allergy relief as well as reducing the pH of colonic contents. The α and ß diversities of the gut microbiota were modulated by L. paracasei AH2 with increased relative abundance of Lacticaseibacillus and SCFA producers (Faecalibaculum, Alloprevotella and Bacteroides genera), as well as decreased unfavorable Lachnospiraceae_NK4A136_group and Alistipes. Additionally, L. paracasei AH2 protected the intestinal barrier function by upregulating tight junctions and improved the antioxidant activities in serum. CONCLUSION: Our findings indicate that L. paracasei AH2 could act as a potential probiotic for relieving wheat allergy by modulating the gut microbiota and elevating SCFA levels. © 2023 Society of Chemical Industry.


Asunto(s)
Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Hipersensibilidad al Trigo , Humanos , Ratones , Animales , Microbioma Gastrointestinal/fisiología , Glútenes , Ratones Endogámicos BALB C , Ácidos Grasos Volátiles
17.
Cells ; 12(24)2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38132109

RESUMEN

Lactic acid bacteria (LAB), a probiotic, provide various health benefits. We recently isolated a new Lactobacillus paracasei strain with strong anti-inflammatory effects under lipopolysaccharide-induced conditions and proposed a new mode of action-augmenting the endoplasmic reticulum stress pathway for anti-inflammatory functions in host cells. The beneficial effects of the L. paracasei strains on the skin have been described; however, the effects of L. paracasei-derived extracellular vesicles (LpEVs) on the skin are poorly understood. Herein, we investigated whether LpEVs can improve inflammation-mediated skin phenotypes by determining their effects on primary human skin cells and a three-dimensional (3D) full-thickness human skin equivalent under tumor necrosis factor (TNF)-α-challenged inflammatory conditions. LpEVs were efficiently taken up by the human skin cells and were much less cytotoxic to host cells than bacterial lysates. Furthermore, low LpEV concentrations efficiently restored TNF-α-induced cellular phenotypes, resulting in increased cell proliferation and collagen synthesis, but decreased inflammatory factor levels (matrix metalloproteinase 1, interleukin 6, and interleukin 8) in the human dermal fibroblasts, which was comparable to that of retinoic acid, a representative antiaging compound. The beneficial effects of LpEVs were validated in a 3D full-thickness human skin equivalent model. LpEV treatment remarkably restored the TNF-α-induced epidermal malformation, abnormal proliferation of keratinocytes in the basal layer, and reduction in dermal collagen synthesis. Additionally, LpEVs penetrated and reached the deepest dermal layer within 24 h when overlaid on top of a 3D full-thickness human skin equivalent. Furthermore, they possessed superior antioxidant capacity compared with the human cell-derived EVs. Taken together, the anti-inflammatory probiotic LpEVs can be attractive antiaging and antioxidant substances for improving inflammation-induced skin phenotypes and disorders.


Asunto(s)
Vesículas Extracelulares , Lacticaseibacillus paracasei , Probióticos , Humanos , Factor de Necrosis Tumoral alfa/metabolismo , Antioxidantes , Probióticos/farmacología , Inflamación , Fenotipo , Antiinflamatorios/farmacología , Vesículas Extracelulares/metabolismo , Colágeno
18.
Foods ; 12(21)2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37959009

RESUMEN

Obesity is a worldwide health problem with a complex interaction between gut microbiota and cognition. Several studies have demonstrated that probiotic treatments improve characteristics linked to obesity. The present study aimed to evaluate the effects of probiotic supplementation on the obesity indexes, inflammatory and oxidative stress markers, gut microbiota, and working memory in obese children. Ten obese children were assigned to receive the probiotics (8 × 109 CFU of Lactobacillus paracasei HII01 and Bifidobacterium animalis subsp. lactis) for 12 weeks. Demographic data were recorded. Urine and fecal samples were collected to evaluate biomarkers related to obesity and cognition. Behavioral working memory was assessed using the visual n-back test. Electroencephalography was employed to measure electrical activity during the visual n-back test. All parameters were evaluated at the baseline and after 12 weeks. The results revealed that probiotic supplementation significantly altered some gut microbial metabolites, gut microbiota, total antioxidant capacity, and neuroinflammatory markers. However, no significant changes were observed in the visual n-back test or electroencephalographic recordings after 12 weeks. In conclusion, the use of probiotics might be an alternative treatment that could improve the gut microbial ecosystem and microbial metabolites, as well as host antioxidant and neuroinflammation levels. The preliminary results indicated that further detailed prolonged studies are needed in order to determine the beneficial effects of the studied probiotics.

19.
Microbiome ; 11(1): 260, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996951

RESUMEN

BACKGROUND: The modulation of immune responses by probiotics is crucial for local and systemic immunity. Recent studies have suggested a correlation between gut microbiota and lung immunity, known as the gut-lung axis. However, the evidence and mechanisms underlying this axis remain elusive. RESULTS: In this study, we screened various Lactobacillus (L.) strains for their ability to augment type I interferon (IFN-I) signaling using an IFN-α/ß reporter cell line. We identified L. paracasei (MI29) from the feces of healthy volunteers, which showed enhanced IFN-I signaling in vitro. Oral administration of the MI29 strain to wild-type B6 mice for 2 weeks resulted in increased expression of IFN-stimulated genes and pro-inflammatory cytokines in the lungs. We found that MI29-treated mice had significantly increased numbers of CD11c+PDCA-1+ plasmacytoid dendritic cells and Ly6Chi monocytes in the lungs compared with control groups. Pre-treatment with MI29 for 2 weeks resulted in less weight loss and lower viral loads in the lung after a sub-lethal dose of influenza virus infection. Interestingly, IFNAR1-/- mice did not show enhanced viral resistance in response to oral MI29 administration. Furthermore, metabolic profiles of MI29-treated mice revealed changes in fatty acid metabolism, with MI29-derived fatty acids contributing to host defense in a Gpr40/120-dependent manner. CONCLUSIONS: These findings suggest that the newly isolated MI29 strain can activate host defense immunity and prevent infections caused by the influenza virus through the gut-lung axis. Video Abstract.


Asunto(s)
Enfermedades Transmisibles , Gripe Humana , Lacticaseibacillus paracasei , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Humanos , Ratones , Animales , Pulmón
20.
Front Bioeng Biotechnol ; 11: 1274020, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901845

RESUMEN

Lactobacillus paracasei has significant potential for development and application in the environmental field, particularly in addressing malodor pollution. This study aims to investigate the cellular response of L. paracasei B1 under high-density culture conditions. The selected strain has previously shown effective deodorizing and bacteriostatic abilities. Transcriptomics techniques are employed to dissect the nutrient metabolism pattern of L. paracasei B1 and its response mechanism under environmental stress. The study characterizes the functions of key differentially expressed genes during growth before and after optimizing the culture conditions. The optimization of fermentation culture conditions provides a suitable growth environment for L. paracasei B1, inducing an enhancement of its phosphotransferase system for sugar source uptake and maintaining high levels of glycolysis and pyruvate metabolism. Consequently, the strain is able to grow and multiply rapidly. Under acid stress conditions, glycolysis and pyruvate metabolism are inhibited, and L. paracasei B1 generates additional energy through aerobic respiration to meet the energy demand. The two-component system and quorum sensing play roles in the response and regulation of L. paracasei B1 to adverse environments. The strain mitigates oxygen stress damage through glutathione metabolism, cysteine and methionine metabolism, base excision repair, and purine and pyrimidine metabolism. Additionally, the strain enhances lysine synthesis, the alanine, aspartate, and glutamate metabolic pathways, and relies on the ABC transport system to accumulate amino acid-compatible solutes to counteract acid stress and osmotic stress during pH regulation. These findings establish a theoretical basis for the further development and application of L. paracasei B1 for its productive properties.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA