Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
1.
Transl Pediatr ; 13(8): 1415-1424, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39263298

RESUMEN

Background: Infantile colic is common in pediatric patients, yet few probiotics effectively treat this condition. The efficacy of Lactobacillus rhamnosus GG (LGG) in managing colic remains unclear. In this meta-analysis, we aimed to evaluate the effectiveness of LGG in treating infantile colic. Methods: We searched PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science databases from their inception until January 2024. We used Version 2 of the Cochrane tool (ROB 2) to assess the risk of bias in randomized trials. Meta-analysis was conducted using RevMan 5.3 software. The inclusion criteria followed the PICOS framework: (I) participants: infants with colic; (II) intervention: LGG administration at any dose; (III) control: placebo or no treatment; (IV) outcomes: primary outcome was crying or fussing time (minutes/day) at the end of the intervention, secondary outcomes included fecal calprotectin content (µg/g) and adverse events; (V) Study type: randomized controlled trials. Results: Four studies involving 168 infants with colic were included. The meta-analysis indicated that LGG significantly reduced daily crying time [mean difference (MD) =-32.59 minutes; 95% confidence interval (CI): -43.23 to -21.96; P<0.001] and fecal calprotectin content (MD =-103.28 µg/g; 95% CI: -149.30 to -7.26; P<0.001). Only one study reported adverse events. Conclusions: LGG is effective in treating infantile colic. Further studies are needed to examine the effects of different doses, administration schedules, and durations of LGG treatment in infants with varying feeding methods.

2.
Int J Biol Macromol ; 278(Pt 3): 134785, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39153668

RESUMEN

Probiotics regulate intestinal flora balance and enhance the intestinal barrier, which is useful in preventing and treating colitis. However, they have strict storage requirements. In addition, they degrade in a strongly acidic environment, resulting in a significant decrease in their activity when used as microbial agents. Lactobacillus rhamnosus GG (LGG) was loaded into acid-resistant and colon-targeting double-layer microgels. The inner layer consists of guar gum (GG) and low methoxyl pectin (LMP), which can achieve retention and degradation in the colon. To achieve colon localization, the outer layer was composed of chitosan (CS) and sodium alginate (SA). The formulation demonstrated favorable bio-responses across various pH conditions in vitro and sustained release of LGG in the colon lesions. Bare LGG survival decreased by 52.2 % in simulated gastric juice (pH 1.2) for 2 h, whereas that of encapsulated LGG decreased by 18.5 %. In the DSS-induced inflammatory model, LGG-loaded microgel significantly alleviated UC symptoms in mice and reduced inflammatory factor levels in the colon. Encapsulation of LGG improved its stability in acidic conditions, thus increasing its content at the colon lesions and reducing pathogenic bacteria. These findings provide an experimental basis and a technical reference for developing and applying probiotic microgel preparations.


Asunto(s)
Alginatos , Quitosano , Colitis Ulcerosa , Lacticaseibacillus rhamnosus , Microgeles , Alginatos/química , Quitosano/química , Animales , Microgeles/química , Ratones , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/terapia , Administración Oral , Probióticos/administración & dosificación , Colon/patología , Colon/microbiología , Colon/metabolismo , Colon/efectos de los fármacos , Galactanos/química , Gomas de Plantas/química , Concentración de Iones de Hidrógeno , Masculino , Modelos Animales de Enfermedad , Sulfato de Dextran , Pectinas/química , Mananos
3.
Biofactors ; 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39135211

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver alterations worldwide, being gut microbiota dysbiosis one of the contributing factors to its development. The aim of this research is to compare the potential effects of a viable probiotic (Lactobacillus rhamnosus GG) with those exerted by its heat-inactivated paraprobiotic counterpart in a dietary rodent model of NAFLD. The probiotic administration effectively prevented the hepatic lipid accumulation induced by a high-fat high-fructose diet feeding, as demonstrated by chemical (lower TG content) and histological (lower steatosis grade and lobular inflammation) analyses. This effect was mainly mediated by the downregulation of lipid uptake (FATP2 protein expression) and upregulating liver TG release to bloodstream (MTTP activity) in rats receiving the probiotic. By contrast, the effect of the paraprobiotic preventing diet-induced liver lipid accumulation was milder, and mainly derived from the downregulation of hepatic de novo lipogenesis (SREBP-1c protein expression and FAS activity) and TG assembly (DGAT2 and AQP9 protein expression). The obtained results demonstrate that under these experimental conditions, the effects induced by the administration of viable L. rhamnosus GG preventing liver lipid accumulation in rats fed a diet rich in saturated fat and fructose differ from those induced by its heat-inactivated paraprobiotic counterpart.

4.
Cureus ; 16(7): e64583, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39144888

RESUMEN

Background Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by intense itching and recurrent eczematous lesions. Important factors in the etiopathogenesis of AD include genetic predisposition, epidermal barrier dysfunction, immune dysregulation, and gut and skin dysbiosis. Probiotics could be a potential preventive strategy for allergies including AD through immune system modulation as well as enhancement of the epithelial barrier integrity. To further understand the role of probiotics in the management of AD, a Knowledge, Attitude, and Practices (KAP) survey was conducted. Materials and methods A steering committee comprising nine experts formulated consensus recommendations on the role of probiotics in the management of AD and associated flare-ups through the use of the Knowledge, Attitude, and Practices questionnaire while analyzing literature reviews and responses from a national panel consisting of 175 members. The evidence strength and quality were evaluated based on the Agency for Healthcare Research and Quality (AHRQ) criteria. The acceptance of expert opinions as recommendations was considered upon receiving an endorsement from ≥70% of the panelists, as indicated by a Likert scale. Results The national panel emphasized that the improvement in nutritional status, immunomodulatory properties, and beneficial effects on the gastrointestinal (GI) tract and skin support the use of probiotics in AD. The panel agreed that probiotics should be a part of the complementary therapy in the management of AD and associated flare-ups. Mostly, a probiotics supplementation duration of eight to 12 weeks is preferred by dermatologists. Probiotics, when used as an adjuvant therapy, may serve as a strategy to reduce steroid usage or maintenance therapy in high-risk cases with flares. Conclusion A Delphi-mediated KAP response provides a real-life approach to the use of probiotics in the management of AD. It suggests that probiotics could be useful as an adjuvant therapy in the management of AD and associated flare-ups when used along with traditional treatment.

5.
J Anim Sci Biotechnol ; 15(1): 89, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38951898

RESUMEN

BACKGROUND: Sows commonly experience insulin resistance in late gestation and lactation, causing lower feed intake and milk production, which can lead to higher mortality rates in newborn piglets. The probiotic Lactobacillus rhamnosus GG (LGG) is known to improve insulin resistance. However, whether supplementing LGG can improve insulin sensitivity in sows and enhance lactation performance, particularly the early survival of offspring remains unclear. Hence, we explored the effects and mechanisms of supplementing LGG during late gestation and lactation on sow insulin sensitivity, lactation performance, and offspring survival. In total, 20 sows were randomly allocated to an LGG (n = 10) and control group (n = 10). RESULTS: In sows, LGG supplementation significantly improved insulin sensitivity during late gestation and lactation, increased feed intake, milk production and colostrum lactose levels in early lactation, and enhanced newborn piglet survival. Moreover, LGG treatment significantly reshaped the gut microbiota in sows, notably increasing microbiota diversity and enriching the relative abundance of insulin sensitivity-associated probiotics such as Lactobacillus, Bifidobacterium, and Bacteroides. Serum metabolite and amino acid profiling in late-gestation sows also revealed decreased branched-chain amino acid and kynurenine serum levels following LGG supplementation. Further analyses highlighted a correlation between mitigated insulin resistance in late pregnancy and lactation by LGG and gut microbiota reshaping and changes in serum amino acid metabolism. Furthermore, maternal LGG enhanced immunity in newborn piglets, reduced inflammation, and facilitated the establishment of a gut microbiota. CONCLUSIONS: We provide the first evidence that LGG mitigates insulin resistance in sows and enhances offspring survival by modulating the gut microbiota and amino acid metabolism.

6.
Imeta ; 3(2): e181, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38882496

RESUMEN

Lactobacillus rhamnosus GG (LGG), the well-characterized human-derived probiotic strain, possesses excellent properties in the maintenance of intestinal homeostasis, immunoregulation and defense against gastrointestinal pathogens in mammals. Here, we demonstrate that the SpaC pilin of LGG causes intestinal epithelium injury by inducing cell pyroptosis and gut microbial dysbiosis in zebrafish. Dietary SpaC activates Caspase-3-GSDMEa pathways in the intestinal epithelium, promotes intestinal pyroptosis and increases lipopolysaccharide (LPS)-producing gut microbes in zebrafish. The increased LPS subsequently activates Gaspy2-GSDMEb pyroptosis pathway. Further analysis reveals the Caspase-3-GSDMEa pyroptosis is initiated by the species-specific recognition of SpaC by TLR4ba, which accounts for the species-specificity of the SpaC-inducing intestinal pyroptosis in zebrafish. The observed pyroptosis-driven gut injury and microbial dysbiosis by LGG in zebrafish suggest that host-specific beneficial/harmful mechanisms are critical safety issues when applying probiotics derived from other host species and need more attention.

7.
Microbiol Res ; 286: 127821, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38941923

RESUMEN

Radiation injury to the intestine is one of the most common complications in patients undergoing abdominal or pelvic cavity radiotherapy. In this study, we investigated the potential protective effect of Lactobacillus rhamnosus GG (LGG) on radiation-induced intestinal injury and its underlying mechanisms. Mice were assigned to a control group, a 10 Gy total abdominal irradiation (TAI) group, or a group pretreated with 108 CFU LGG for three days before TAI. Small intestine and gut microbiota were analyzed 3.5 days post-exposure. LGG intervention improved intestinal structure, reduced jejunal DNA damage, and inhibited the inflammatory cGAS/STING pathway. Furthermore, LGG reduced M1 proinflammatory macrophage and CD8+ T cell infiltration, restoring the balance between Th17 and Treg cells in the inflamed jejunum. LGG also partially restored the gut microbiota. These findings suggest the possible therapeutic radioprotective effect of probiotics LGG in alleviating radiation-induced intestinal injury by maintaining immune homeostasis and reshaping gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Probióticos , Animales , Microbioma Gastrointestinal/efectos de la radiación , Ratones , Probióticos/administración & dosificación , Traumatismos por Radiación/inmunología , Macrófagos/inmunología , Intestinos/microbiología , Intestinos/efectos de la radiación , Intestinos/inmunología , Daño del ADN , Linfocitos T CD8-positivos/inmunología , Proteínas de la Membrana/metabolismo , Linfocitos T Reguladores/inmunología , Masculino , Células Th17/inmunología , Yeyuno/efectos de la radiación , Yeyuno/inmunología , Yeyuno/microbiología , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/prevención & control , Nucleotidiltransferasas
8.
PeerJ ; 12: e17427, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827289

RESUMEN

Background: Survivors of sepsis may encounter cognitive impairment following their recovery from critical condition. At present, there is no standardized treatment for addressing sepsis-associated encephalopathy. Lactobacillus rhamnosus GG (LGG) is a prevalent bacterium found in the gut microbiota and is an active component of probiotic supplements. LGG has demonstrated to be associated with cognitive improvement. This study explored whether LGG administration prior to and following induced sepsis could ameliorate cognitive deficits, and explored potential mechanisms. Methods: Female C57BL/6 mice were randomly divided into three groups: sham surgery, cecal ligation and puncture (CLP), and CLP+LGG. Cognitive behavior was assessed longitudinally at 7-9d, 14-16d, and 21-23d after surgery using an open field test and novel object recognition test. The impact of LGG treatment on pathological changes, the expression level of brain-derived neurotrophic factor (BDNF), and the phosphorylation level of the TrkB receptor (p-TrkB) in the hippocampus of mice at two weeks post-CLP (16d) were evaluated using histological, immunofluorescence, immunohistochemistry, and western blot analyses. Results: The CLP surgery induced and sustained cognitive impairment in mice with sepsis for a minimum of three weeks following the surgery. Compared to mice subjected to CLP alone, the administration of LGG improved the survival of mice with sepsis and notably enhanced their cognitive functioning. Moreover, LGG supplementation significantly alleviated the decrease in hippocampal BDNF expression and p-TrkB phosphorylation levels caused by sepsis, preserving neuronal survival and mitigating the pathological changes within the hippocampus of mice with sepsis. LGG supplementation mitigates sepsis-related cognitive impairment in mice and preserves BDNF expression and p-TrkB levels in the hippocampus.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Disfunción Cognitiva , Hipocampo , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Probióticos , Sepsis , Animales , Sepsis/complicaciones , Sepsis/terapia , Sepsis/microbiología , Sepsis/metabolismo , Disfunción Cognitiva/terapia , Disfunción Cognitiva/etiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Ratones , Hipocampo/metabolismo , Probióticos/farmacología , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Modelos Animales de Enfermedad , Receptor trkB/metabolismo , Encefalopatía Asociada a la Sepsis/metabolismo , Encefalopatía Asociada a la Sepsis/patología , Encefalopatía Asociada a la Sepsis/dietoterapia , Fosforilación
9.
Pharmacol Res ; 206: 107275, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908615

RESUMEN

Triptolide (TP) is the principal bioactive compound of Tripterygium wilfordii with significant anti-tumor, anti-inflammatory and immunosuppressive activities. However, its severe hepatotoxicity greatly limits its clinical use. The underlying mechanism of TP-induced liver damage is still poorly understood. Here, we estimate the role of the gut microbiota in TP hepatotoxicity and investigate the bile acid metabolism mechanisms involved. The results of the antibiotic cocktail (ABX) and fecal microbiota transplantation (FMT) experiment demonstrate the involvement of intestinal flora in TP hepatotoxicity. Moreover, TP treatment significantly perturbed gut microbial composition and reduced the relative abundances of Lactobacillus rhamnosus GG (LGG). Supplementation with LGG reversed TP-induced hepatotoxicity by increasing bile salt hydrolase (BSH) activity and reducing the increased conjugated bile acids (BA). LGG supplementation upregulates hepatic FXR expression and inhibits NLRP3 inflammasome activation in TP-treated mice. In summary, this study found that gut microbiota is involved in TP hepatotoxicity. LGG supplementation protects mice against TP-induced liver damage. The underlying mechanism was associated with the gut microbiota-BA-FXR axis. Therefore, LGG holds the potential to prevent and treat TP hepatotoxicity in the clinic.


Asunto(s)
Ácidos y Sales Biliares , Enfermedad Hepática Inducida por Sustancias y Drogas , Diterpenos , Compuestos Epoxi , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Fenantrenos , Receptores Citoplasmáticos y Nucleares , Animales , Diterpenos/farmacología , Fenantrenos/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Compuestos Epoxi/farmacología , Ácidos y Sales Biliares/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Probióticos/uso terapéutico , Probióticos/farmacología , Trasplante de Microbiota Fecal , Inflamasomas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Front Cell Infect Microbiol ; 14: 1371916, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38716199

RESUMEN

Porcine epidemic diarrhea virus (PEDV) has become a challenging problem in pig industry worldwide, causing significant profit losses. Lactobacillus rhamnosus GG (LGG) has been regarded as a safe probiotic strain and has been shown to exert protective effects on the intestinal dysfunction caused by PEDV. This study evaluated the effect of LGG on the gut health of lactating piglets challenged with PEDV. Fifteen piglets at 7 days of age were equally assigned into 3 groups (5 piglets per group): 1) control group (basal diet); 2) PEDV group: (basal diet + PEDV challenged); 3) LGG + PEDV group (basal diet + 3×109 CFU/pig/day LGG + PEDV). The trial lasted 11 days including 3 days of adaptation. The treatment with LGG was from D4 to D10. PEDV challenge was carried out on D8. PEDV infection disrupted the cell structure, undermined the integrity of the intestinal tract, and induced oxidative stress, and intestinal damage of piglets. Supplementation of LGG improved intestinal morphology, enhanced intestinal antioxidant capacity, and alleviated jejunal mucosal inflammation and lipid metabolism disorders in PEDV-infected piglets, which may be regulated by LGG by altering the expression of TNF signaling pathway, PPAR signaling pathway, and fat digestion and absorption pathway.


Asunto(s)
Infecciones por Coronavirus , Suplementos Dietéticos , Lacticaseibacillus rhamnosus , Virus de la Diarrea Epidémica Porcina , Probióticos , Enfermedades de los Porcinos , Animales , Porcinos , Probióticos/administración & dosificación , Enfermedades de los Porcinos/prevención & control , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/terapia , Estrés Oxidativo , Intestinos/patología , Polvos , Mucosa Intestinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA