Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 446
Filter
1.
Adv Sci (Weinh) ; : e2309725, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38647360

ABSTRACT

The interplay between bacteria and their host influences the homeostasis of the human immune microenvironment, and this reciprocal interaction also affects the process of tissue damage repair. A variety of immunomodulatory commensal bacteria reside in the body, capable of delivering membrane vesicles (MVs) to host cells to regulate the local immune microenvironment. This research revealed, for the initial time, the significant enhancement of mucosal and cutaneous wound healing by MVs secreted by the human commensal Lactobacillus reuteri (RMVs) through modulation of the inflammatory environment in wound tissue. Local administration of RMVs reduces the proportion of pro-inflammatory macrophages in inflamed tissues and mitigates the level of local inflammation, thereby facilitating the healing of oral mucosa and cutaneous wounds. The elevated oxidative stress levels in activated pro-inflammatory macrophages can be modulated by RMVs, resulting in phenotypic transformation of macrophages. Furthermore, 3-hydroxypropionaldehyde present in RMVs can decrease the mitochondrial permeability of macrophages and stabilize the mitochondrial membrane potential, thereby promoting the conversion of macrophages to an anti-inflammatory phenotype. This study pioneers the significance of commensal bacterial MVs in tissue injury repair and presents a novel concept for the repair of tissue damage.

2.
Biol Direct ; 19(1): 23, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38500127

ABSTRACT

BACKGROUND: This study seeks to investigate the impacts of Lactobacillus reuteri (L. reuteri) on hepatic ischemia-reperfusion (I/R) injury and uncover the mechanisms involved. METHODS: Mice in the I/R groups were orally administered low and high doses of L.reuteri (L.reuteri-low and L. reuteri-hi; 1 × 1010 CFU/d and 1 × 1011 CFU/d), for 4 weeks prior to surgery. Following this, mice in the model group were treated with an Nrf2 inhibitor (ML-385), palmitoylcarnitine, or a combination of both. RESULTS: After treatment with L. reuteri, mice exhibited reduced levels of serum aminotransferase (ALT), aspartate aminotransferase (AST), and myeloperoxidase (MPO) activity, as well as a lower Suzuki score and apoptosis rate. L. reuteri effectively reversed the I/R-induced decrease in Bcl2 expression, and the significant increases in the levels of Bax, cleaved-Caspase3, p-p65/p65, p-IκB/IκB, p-p38/p38, p-JNK/JNK, and p-ERK/ERK. Furthermore, the administration of L. reuteri markedly reduced the inflammatory response and oxidative stress triggered by I/R. This treatment also facilitated the activation of the Nrf2/HO-1 pathway. L. reuteri effectively counteracted the decrease in levels of beneficial gut microbiota species (such as Blautia, Lachnospiraceae NK4A136, and Muribaculum) and metabolites (including palmitoylcarnitine) induced by I/R. Likewise, the introduction of exogenous palmitoylcarnitine demonstrated a beneficial impact in mitigating hepatic injury induced by I/R. However, when ML-385 was administered prior to palmitoylcarnitine treatment, the previously observed effects were reversed. CONCLUSION: L. reuteri exerts protective effects against I/R-induced hepatic injury, and its mechanism may be related to the promotion of probiotic enrichment, differential metabolite homeostasis, and the Nrf2/HO-1 pathway, laying the foundation for future clinical applications.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Reperfusion Injury , Mice , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/therapeutic use , Palmitoylcarnitine/therapeutic use , Reperfusion Injury/prevention & control , Reperfusion Injury/drug therapy , Ischemia
3.
Adv Sci (Weinh) ; : e2307233, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38487926

ABSTRACT

The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.

4.
Int J Biol Macromol ; 261(Pt 2): 129733, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38307433

ABSTRACT

The function of miRNAs in intestinal inflammatory injury regulation has been studied extensively. However, the targeted delivery of these functional nucleic acid molecules to specific organs through encapsulation carriers and exerting their functional effects remain critical challenges for further research. Here, we constructed miR-146a-5p overexpression plasmid and validated the anti-inflammatory properties in the cell model. Then, the plasmid was encapsulated by the Pickering double emulsion system to investigate the role of Pickering double emulsion system in LPS-induced acute intestinal inflammatory injury. The results showed that the Pickering double emulsion system could effectively protect the integrity of plasmids in the intestinal tract, alleviate intestinal inflammatory injury, and upregulate the relative abundance of Lactobacillus reuteri. Mechanically, in vivo and in vitro experiments have shown that miR-146a-5p inhibits TLR4/NF-κB pathway to alleviate intestinal inflammation. In addition, miR-146a-5p can also regulate intestinal homeostasis by targeting the RNA polymerase sigma factor RpoD and α-galactosidase A, thereby affecting the growth of Lactobacillus reuteri. Above all, this study reveals a potential mechanism for miR-146a-5p to treat intestinal inflammation and provides a new delivery strategy for miRNAs to regulate intestinal homeostasis.


Subject(s)
Gastrointestinal Microbiome , MicroRNAs , Humans , Emulsions , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Inflammation/drug therapy , Inflammation/genetics
5.
Int J Biol Macromol ; 262(Pt 2): 130152, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38365143

ABSTRACT

Supplementing probiotics or indigestible carbohydrates is a usual strategy to prevent or revert unhealthy states of the gut by reshaping gut microbiota. One criterion that probiotics are efficacious is the capacity to survive in the gastrointestinal tract. Biofilm is the common growth mode of microorganisms with high tolerances toward harsh environments. Suitable scaffolds are crucial for successful biofilm culture and large-scale production of biofilm-phenotype probiotics. However, the role of scaffolds containing indigestible carbohydrates in biofilm formation has not been studied. In this study, porous zein/cellulose composite scaffolds provided nitrogen sources and carbon sources simultaneously at the solid/liquid interfaces, being beneficial to the biofilm formation of Lactobacillus reuteri. The biofilms showed 2.1-17.4 times higher tolerances in different gastrointestinal conditions. In human fecal fermentation, the biofilms combined with the zein/cellulose composite scaffolds act as the "synbiotics" positively modulating the gut microbiota and the short-chain fatty acids (SCFAs), where biofilms provide probiotics and scaffolds provide prebiotics. The "synbiotics" show a more positive regulation ability than planktonic L. reuteri, presenting potential applications in gut health interventions. These results provide an understanding of the synergistic effects of biofilm-phenotype probiotics and indigestible carbohydrates contained in the "synbiotics" in gut microbiota modulation.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Synbiotics , Zein , Humans , Cellulose , Porosity , Prebiotics , Carbohydrates , Biofilms
6.
Gut Microbes ; 16(1): 2313769, 2024.
Article in English | MEDLINE | ID: mdl-38353638

ABSTRACT

Melatonin has various physiological effects, such as the maintenance of circadian rhythms, anti-inflammatory functions, and regulation of intestinal barriers. The regulatory functions of melatonin in gut microbiota remodeling have also been well clarified; however, the role of gut microbiota in regulating host melatonin production remains poorly understood. To address this, we studied the contribution of gut microbiota to host melatonin production using gut microbiota-perturbed models. We demonstrated that antibiotic-treated and germ-free mice possessed diminished melatonin levels in the serum and elevated melatonin levels in the colon. The influence of the intestinal microbiota on host melatonin production was further confirmed by fecal microbiota transplantation. Notably, Lactobacillus reuteri (L. R) and Escherichia coli (E. coli) recapitulated the effects of gut microbiota on host melatonin production. Mechanistically, L. R and E. coli activated the TLR2/4/MyD88/NF-κB signaling pathway to promote expression of arylalkylamine N-acetyltransferase (AANAT, a rate-limiting enzyme for melatonin production), and MyD88 deficiency in colonic epithelial cells abolished the influence of intestinal microbiota on colonic melatonin production. Collectively, we revealed a specific underlying mechanism of gut microbiota to modulate host melatonin production, which might provide novel therapeutic ideas for melatonin-related diseases.


Subject(s)
Gastrointestinal Microbiome , Melatonin , Animals , Mice , Escherichia coli , Myeloid Differentiation Factor 88/genetics , Adaptor Proteins, Signal Transducing , Epithelial Cells
7.
Microb Pathog ; 188: 106541, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38224920

ABSTRACT

Diarrhea is a prevalent health issue in farm animals and poses a significant challenge to the progress of animal husbandry. Recent evidence suggested that probiotics can alleviate diarrhea by maintaining gut microbial balance and enhancing the integrity of the intestinal barrier. However, there is a scarcity of studies investigating the efficacy of equine Lactobacillus reuteri in relieving E. coli-induced diarrhea. Hence, this study aimed to examine the potential of equine-derived Lactobacillus reuteri in alleviating E. coli diarrhea from the perspective of gut microbiota. Results demonstrated that supplementation of Lactobacillus reuteri had the potential to alleviate diarrhea induced by E. coli infection and restore the decline of tight junction genes, such as Claudin-1 and ZO-1. Additionally, Lactobacillus reuteri supplementation can restore the expression of inflammatory factors (IL-6, IL-10, TNF-α, and IFN-γ) and reduce colon inflammatory damage. Diversity analysis, based on amplicon sequencing, revealed a significant reduction in the diversity of gut microbiota during E. coli-induced diarrhea. Moreover, there were notable statistical differences in the composition and structure of gut microbiota among the different treatment groups. E. coli could induce gut microbial dysbiosis by decreasing the abundance of beneficial bacteria, including Lactobacillus, Bifidobacterium, Ligilactobacillus, Enterorhabdus, and Lachnospiraceae_UCG_001, in comparison to the control group. Conversely, supplementation with Lactobacillus reuteri could restore the abundance of beneficial bacteria and increase the diversity of the gut microbiota, thereby reshaping gut microbiota. Additionally, we also observed that supplementation with Lactobacillus reuteri alone improved the gut microbial composition and structure. In summary, the findings suggest that Lactobacillus reuteri can alleviate E. coli-induced diarrhea by preserving the integrity of the intestinal barrier and modulating the composition of the gut microbiota. These results not only contribute to understanding of the mechanism underlying the beneficial effects of Lactobacillus reuteri in relieving diarrhea, but also provide valuable insights for the development of probiotic products aimed at alleviating diarrheal diseases.


Subject(s)
Escherichia coli Infections , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Horses , Animals , Escherichia coli , Diarrhea/therapy , Lactobacillus , Escherichia coli Infections/therapy , Escherichia coli Infections/veterinary , Probiotics/therapeutic use , Probiotics/pharmacology
8.
Mol Nutr Food Res ; 68(2): e2300337, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38048544

ABSTRACT

SCORE: This study identifies the coding gene (aldB) of acetolactate decarboxylase (ALDC) as an important regulatory gene of the intracellular pH in Lactobacillus reuteri (L. reuteri), uncovering the important role of ALDC in regulating intracellular pH, morphological features, and antagonism properties in the probiotic organism L. reuteri. METHODS AND RESULTS: The aldB mutant (ΔaldB) of L. reuteri is established using the homologous recombination method. Compare to the wild-type (WT) strain, the ΔaldB strain shows a smaller body size, grows more slowly, and contains more acid in the cell cytoplasm. The survival rate of the ΔaldB strain is much lower in low pH and simulated gastric fluid (SGF) than that of the WT strain, but higher in simulated intestinal fluid (SIF). The antagonism test demonstrates the ΔaldB strain can inhibit Listeria monocytogenes (L. monocytogenes) and Salmonella more effectively than the WT strain. Additionally, there is a dramatic decrease in the adhesion rate of Salmonella to Caco-2 and HT-29 cells in the presence of the ΔaldB strain compared to the WT strain. Simultaneously analyze, the auto-aggregation, co-aggregation, cell surface hydrophobicity (CSH), hemolytic, temperature, NaCl, oxidative stress, and antibiotic susceptibility of the ΔaldB strain are consistent with the features of probiotics. CONCLUSION: This study highlights that the aldB gene plays a significant role in the growth and antibacterial properties of L. reuteri.


Subject(s)
Carboxy-Lyases , Limosilactobacillus reuteri , Probiotics , Humans , Caco-2 Cells , Probiotics/pharmacology , Hydrogen-Ion Concentration
9.
Lett Appl Microbiol ; 77(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38126116

ABSTRACT

Fecal microbiota transplantation from patients with depression/inflammatory bowel disease (PDI) causes depression with gut inflammation in mice. Here, we investigated the effects of six Lactobacillus reuteri strains on brain-derived neurotropic factor (BDNF), serotonin, and interleukin (IL)-6 expression in neuronal or macrophage cells and PDI fecal microbiota-cultured microbiota (PcM)-induced depression in mice. Of these strains, L6 most potently increased BDNF and serotonin levels in corticosterone-stimulated SH-SY5Y and PC12 cells, followed by L3. L6 most potently decreased IL-6 expression in lipopolysaccharide (LPS)-stimulated macrophages. When L1 (weakest in vitro), L3, and L6 were orally administered in mice with PcM-induced depression, L6 most potently suppressed depression-like behaviors and hippocampal TNF-α and IL-6 expression and increased hippocampal serotonin, BDNF, 5HT7, GABAARα1, and GABABR1b expression, followed by L3 and L1. L6 also suppressed TNF-α and IL-6 expression in the colon. BDNF or serotonin levels in corticosterone-stimulated neuronal cells were negatively correlated with depression-related biomarkers in PcM-transplanted mice, while IL-6 levels in LPS-stimulated macrophage were positively correlated. These findings suggest that IL-6 expression-suppressing and BDNF/serotonin expression-inducing LBPs in vitro, particularly L6, may alleviate gut microbiota-involved depression with colitis in vivo.


Subject(s)
Gastrointestinal Microbiome , Limosilactobacillus reuteri , Neuroblastoma , Rats , Humans , Mice , Animals , Interleukin-6/genetics , Depression/therapy , Tumor Necrosis Factor-alpha/genetics , Lipopolysaccharides/toxicity , Corticosterone/pharmacology , Serotonin/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Anxiety/therapy , Anxiety/etiology , Mice, Inbred C57BL
10.
Biomedicines ; 11(12)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38137342

ABSTRACT

This study aimed to elucidate the unique chemical compositions of plasma-activated water (PAW) and the potential antibacterial efficacy of PAW as a novel vaginal cleanser. We analyzed the ion compositions (four anions: F-, Cl-, NO3-, SO42-; five cations: Na+, NH4+, K+, Mg2+, Ca2+) of several formulations of PAW generated at different electrical powers (12 and 24 V) at various treatment time points (1, 10, and 20 min), and stay durations (immediate, 30, and 60 min). As treatment duration increased, hypochlorous acid (HOCl), Ca2+, and Mg2+ concentrations increased and Cl- concentration decreased. Higher electrical power and longer treatment duration resulted in increased HOCl levels, which acts to prevent the growth of general microorganisms. Notably, PAW had no antibacterial effects against the probiotic, Lactobacillus reuteri, which produces lactic acid and is important for vaginal health. These findings indicate that PAW contains HOCl and some cations (Ca2+ and Mg2+), which should help protect against pathogens of the vaginal mucosa and have a cleansing effect within the vaginal environment while not harming beneficial bacteria.

12.
Exp Neurobiol ; 32(5): 313-327, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37927130

ABSTRACT

Mental health is influenced by the gut-brain axis; for example, gut dysbiosis has been observed in patients with major depressive disorder (MDD). Gut microbial changes by fecal microbiota transplantation or probiotics treatment reportedly modulates depressive symptoms. However, it remains unclear how gut dysbiosis contributes to mental dysfunction, and how correction of the gut microbiota alleviates neuropsychiatric disorders. Our previous study showed that chronic consumption of Lactobacillus reuteri ATG-F4 (F4) induced neurometabolic alterations in healthy mice. Here, we investigated whether F4 exerted therapeutic effects on depressive-like behavior by influencing the central nervous system. Using chronic unpredictable stress (CUS) to induce anhedonia, a key symptom of MDD, we found that chronic F4 consumption alleviated CUS-induced anhedonic behaviors, accompanied by biochemical changes in the gut, serum, and brain. Serum and brain metabolite concentrations involved in tryptophan metabolism were regulated by CUS and F4. F4 consumption reduced the elevated levels of serotonin (5-HT) in the brain observed in the CUS group. Additionally, the increased expression of Htr1a, a subtype of the 5-HT receptor, in the medial prefrontal cortex (mPFC) of stressed mice was restored to levels observed in stress-naïve mice following F4 supplementation. We further demonstrated the role of Htr1a using AAV-shRNA to downregulate Htr1a in the mPFC of CUS mice, effectively reversing CUS-induced anhedonic behavior. Together, our findings suggest F4 as a potential therapeutic approach for relieving some depressive symptoms and highlight the involvement of the tryptophan metabolism in mitigating CUS-induced depressive-like behaviors through the action of this bacterium.

13.
J Agric Food Chem ; 71(48): 19101-19110, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37988599

ABSTRACT

There is an increasing global demand for probiotics because of their numerous health benefits. However, a significant percentage of commercially available probiotic products have microbial quantities that are not in accordance with their product labels. In quantifying bacteria, the viable plate count is the standard method but is considered laborious and time-consuming. We demonstrate the use of an attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy coupled with chemometrics to quantify a pure culture of Lactobacillus reuteri (L. reuteri) ProTectis grown in deMan, Rogosa, and Sharpe broth. The chemometric partial least-squares model generated was able to accurately quantify viable plate count (VPC) (root-mean-square error of cross-validation (RMSECV) = 0.115 log CFU mL-1, root-mean-square error of prediction (RMSEP) = 0.145 log CFU mL-1, R2 = 0.982). These results provide proof of concept for this quantification technique and can potentially be developed and applied for the quantification of L. reuteri ProTectis in food products.


Subject(s)
Limosilactobacillus reuteri , Spectroscopy, Fourier Transform Infrared/methods , Chemometrics , Fourier Analysis , Least-Squares Analysis
14.
BMC Cancer ; 23(1): 1044, 2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37904102

ABSTRACT

BACKGROUND: Pancreatic cancer is a highly lethal disease with no effective treatments. Lactobacillus casei (L. casei) and Lactobacillus reuteri (L. reuteri) exhibited therapeutic effects on several cancers, but their roles in pancreatic cancer are unknown. This study aims to explore how L. casei & L. reuteri influence pancreatic cancer and the underlying mechanisms. METHODS: Pancreatic cancer cells were treated with L. casei & L. reuteri and co-cultured with macrophages in a transwell system in vitro. Pancreatic cancer xenograft model was established and L. casei & L. reuteri was used to treat mice in vivo. MTT, CCK-8 assay or immunohistochemical staining were used to determine the proliferation of pancreatic cancer cells or tumor tissues. Transwell assay was applied to test the migration and invasion of pancreatic cells. RT-qPCR was utilized to assess TLR4 and MyD88 expressions in pancreatic cells or tumor tissues. WB, immunofluorescence staining, or flow cytometry was used to evaluate the M1/M2 polarization of macrophages. Besides, the composition of gut microbiota of tumor-bearing mice was determined by 16 S rRNA sequencing, and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) untargeted metabolomics was used to evaluate the metabolic profiles of feces. RESULTS: L. casei & L. reuteri inhibited the proliferation, migration, invasion of pancreatic cancer cells and pancreatic cancer cell-induced M2 polarization of macrophages by suppressing TLR4. Meanwhile, L. casei & L. reuteri repressed pancreatic cancer growth and promoted M1 macrophage polarization. Besides, L. casei & L. reuteri reduced fecal Alloprevotella and increased fecal azelate and glutamate in nude mice, while TLR4 inhibitor TAK-242 increased Clostridia UCG-014, azelate, uridine, methionine sulfoxide, oxypurinol, and decreased glyceryl monoester in the feces of pancreatic tumor-bearing mice. Fecal oxypurinol and glyceryl monoester levels were positively or negatively associated with gut Clostridia UCG-014 abundance, respectively. CONCLUSION: L. casei & L. reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis.


Subject(s)
Gastrointestinal Microbiome , Lacticaseibacillus casei , Limosilactobacillus reuteri , Pancreatic Neoplasms , Mice , Humans , Animals , Toll-Like Receptor 4/metabolism , Mice, Nude , Chromatography, Liquid , Oxypurinol/metabolism , Oxypurinol/pharmacology , Tandem Mass Spectrometry , Macrophages/metabolism , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms
15.
Nutrients ; 15(19)2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37836540

ABSTRACT

Acute lung injury (ALI) causes lung inflammation and edema as well as resulting in gut microbiota disorder. Probiotics, however, can improve the gut microbiota composition and modulate its immune response, playing an important role in ALI pathogenesis. Therefore, our study aims to investigate the effect of Lactobacillus reuteri on Lipopolysaccharide (LPS)-induced ALI in mice and to probe the mechanism of its synergistic modulatory effect on the lungs and intestines. We assessed the therapeutic effects of L. reuteri in the ALI mouse model by histopathology, alveolar lavage fluid and serum inflammatory factor analysis and explored microbiome and transcriptome alterations. L. reuteri intervention effectively attenuated lung tissue injury and significantly reduced the LPS-induced inflammatory response and macrophage and neutrophil infiltration. Additionally, L. reuteri improved the intestinal barrier function and remodeled the disordered microbiota. In conclusion, our study showed that L. reuteri attenuated the inflammatory response, ameliorated the pulmonary edema, repaired the intestinal barrier, and remodeled the gut microbiota in ALI mice. This study provides new perspectives on the clinical treatment of ALI.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Animals , Mice , Lipopolysaccharides/adverse effects , Acute Lung Injury/chemically induced , Acute Lung Injury/therapy , Acute Lung Injury/pathology , Lung/pathology
16.
Front Cell Infect Microbiol ; 13: 1254198, 2023.
Article in English | MEDLINE | ID: mdl-37662007

ABSTRACT

Objectives: Digestive system diseases have evolved into a growing global burden without sufficient therapeutic measures. Lactobacillus reuteri (L. reuteri) is considered as a new potential economical therapy for its probiotic effects in the gastrointestinal system. We have provided an overview of the researches supporting various L. reuteri strains' application in treating common digestive system diseases, including infantile colic, diarrhea, constipation, functional abdominal pain, Helicobacter pylori infection, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases. Methods: The summarized literature in this review was derived from databases including PubMed, Web of Science, and Google Scholar. Results: The therapeutic effects of L. reuteri in digestive system diseases may depend on various direct and indirect mechanisms, including metabolite production as well as modulation of the intestinal microbiome, preservation of the gut barrier function, and regulation of the host immune system. These actions are largely strain-specific and depend on the activation or inhibition of various certain signal pathways. It is well evidenced that L. reuteri can be effective both as a prophylactic measure and as a preferred therapy for infantile colic, and it can also be recommended as an adjuvant strategy to diarrhea, constipation, Helicobacter pylori infection in therapeutic settings. While preclinical studies have shown the probiotic potential of L. reuteri in the management of functional abdominal pain, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases, its application in these disease settings still needs further study. Conclusion: This review focuses on the probiotic effects of L. reuteri on gut homeostasis via certain signaling pathways, and emphasizes the importance of these probiotics as a prospective treatment against several digestive system diseases.


Subject(s)
Colic , Colorectal Neoplasms , Digestive System Diseases , Diverticulitis , Helicobacter Infections , Helicobacter pylori , Inflammatory Bowel Diseases , Limosilactobacillus reuteri , Humans , Helicobacter Infections/therapy , Digestive System Diseases/therapy , Constipation , Abdominal Pain , Diarrhea
17.
Pediatr Neonatol ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37684161

ABSTRACT

BACKGROUND: Group B Streptococci (GBS) are common vaginal bacteria found in 20-30% of pregnant women and a significant cause of invasive infections in newborns. Recently, attention has been focused on the efficacy of probiotics during the perinatal period. However, the effect of probiotic intake on the mother-to-child transmission (MTCT) of GBS remains unknown. METHODS: Pregnant women with positive GBS results from vaginal and rectal swab cultures at 35-37 weeks of gestation were randomly assigned to the probiotic group or the control group in an open-label manner at the Department of Obstetrics and Gynecology, San-ikukai Hospital, Tokyo, Japan. The probiotic group received Lactobacillus reuteri during antenatal checkups from 35 to 37-week gestation to 1 month after delivery. Rectal swabs were obtained from the newborns at 5 days and at 1 month of age. Whole-genome sequencing was performed to test for GBS strains in the mother, whose newborn carried GBS at the 1-month checkup. Multi-locus sequence typing and single nucleotide polymorphism analyses were performed to identify MTCT. RESULTS: Overall, 67 mother-infant pairs were included, with 31 in the probiotic group and 36 in the control group. The positivity rate of GBS in newborns at 1 month of age was 10% (n = 3) in the probiotic group and 28% (n = 10) in the control group. In newborns carrying GBS at 1 month of age, genetic analysis revealed that the MTCT rate was 6% in the probiotic group and 22% in the control group, although the difference was not statistically significant (p = 0.0927). CONCLUSION: No statistically significant difference was found; however, the consumption of L. reuteri by women with GBS-positive pregnancies may inhibit the MTCT of GBS.

18.
BMC Complement Med Ther ; 23(1): 340, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37752485

ABSTRACT

BACKGROUND: The development of new strategies to inhibit and/or treat osteoporosis as a chronic systemic disease is one of the most crucial topics. The present study aimed to investigate the simultaneous effects of calcium fluoride nanoparticles (CaF2 NPs) and lactobacillus reuteri ATCC PTA 6475 (L. reuteri) against osteoporosis in an ovariectomized rat model (OVX). METHODS: In this study, 18 matured Wistar female rats were randomly assigned into 6 groups, including control, OVX, sham, OVX + L. reuteri, OVX + CaF2 NPs, and OVX + L. reuteri + CaF2 NPs. We used OVX rats to simulate post-menopausal osteoporosis, and the treatments were begun two weeks before OVX and continued for four weeks. All groups' blood samples were collected, and serum biomarkers (estrogen, calcium, vitamin D3, and alkaline phosphatase (ALP)) were measured. The tibia and Femur lengths of all groups were measured. Histopathological slides of tibia, kidney, and liver tissues were analyzed using the Hematoxylin and Eosin staining method. RESULTS: Our results revealed that dietary supplementation of L. reuteri and CaF2 NPs in low doses for 6 weeks did not show adverse effects in kidney and liver tissues. The tibial and femoral lengths of OVX rats as well as the population of osteoblasts and osteocytes and newly generated osteoid in the tibia remarkably increased in the combination therapy group. Moreover, there was a significant increase in serum estrogen levels and a significant decrease in serum calcium and alkaline phosphatase levels in combination treatment groups compared to the OVX groups not receiving the diet. CONCLUSIONS: Our results suggest the favorable effects of the simultaneous supplementation of L. reuteri and CaF2 NP to reduce post-menopausal bone loss.


Subject(s)
Limosilactobacillus reuteri , Osteoporosis, Postmenopausal , Osteoporosis , Female , Animals , Rats , Humans , Rats, Wistar , Calcium Fluoride , Alkaline Phosphatase , Calcium , Osteoporosis/drug therapy , Estrogens , Dietary Supplements
19.
Gut Microbes ; 15(2): 2256043, 2023 12.
Article in English | MEDLINE | ID: mdl-37698879

ABSTRACT

Intestinal microbes impact the health of the intestine and organs distal to the gut. Limosilactobacillus reuteri is a human intestinal microbe that promotes normal gut transit, the anti-inflammatory immune system, wound healing, normal social behavior in mice, and prevents bone reabsorption. Oxytocin impacts these functions and oxytocin signaling is required for L. reuteri-mediated wound healing and social behavior; however, the events in the gut leading to oxytocin stimulation and beneficial effects are unknown. Here we report evolutionarily conserved oxytocin production in the intestinal epithelium through analysis of single-cell RNA-Seq datasets and imaging of human and mouse intestinal tissues. Moreover, human intestinal organoids produce oxytocin, demonstrating that the intestinal epithelium is sufficient to produce oxytocin. We find that L. reuteri facilitates oxytocin secretion from human intestinal tissue and human intestinal organoids. Finally, we demonstrate that stimulation of oxytocin secretion by L. reuteri is dependent on the gut hormone secretin, which is produced in enteroendocrine cells, while oxytocin itself is produced in enterocytes. Altogether, this work demonstrates that oxytocin is produced and secreted from enterocytes in the intestinal epithelium in response to secretin stimulated by L. reuteri. This work thereby identifies oxytocin as an intestinal hormone and provides mechanistic insight into avenues by which gut microbes promote host health.


Subject(s)
Gastrointestinal Hormones , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Humans , Animals , Mice , Secretin , Oxytocin , Intestinal Mucosa
20.
J Indian Soc Periodontol ; 27(4): 352-361, 2023.
Article in English | MEDLINE | ID: mdl-37593565

ABSTRACT

Peri-implant diseases are prevalent conditions, but a predictable management strategy is still lacking. The objective of the present article was to evaluate the adjunctive benefits of probiotics with nonsurgical therapy in the management of peri-implant diseases. The review protocol was registered in PROSPERO and prepared according to PRISMA guidelines. Randomized controlled clinical trials in patients diagnosed with the peri-implant disease where probiotic was used as an adjunct to nonsurgical therapy were included in the study. The risk difference of percentage reduction in bleeding on probing, plaque accumulation, and mean difference in probing pocket depth reductions at implant level were estimated using a random effect model due to high heterogeneity among studies. Four studies fulfilled the criteria for selection. Two of them presented data on both peri-implantitis and peri-implant mucositis and they were considered separate studies during meta-analysis. Significant reduction in percentage of bleeding on probing was noticed at 1 and 3 months (-0.28 [-0.48, -0.09], P = 0.004 and - 0.19 [-0.35, -0.02], P = 0.03, respectively), but the reduction was not statistically significant at 6 months. Similar results were also observed for plaque accumulation. No statistically significant reduction in probing pocket depth was observed in the probiotic group during any of the re-evaluations. Conclusion: Adjunctive therapy of probiotics may improve the efficacy of nonsurgical therapy of peri-implant diseases for up to 3 months. However, moderate certainty was observed for a reduction in bleeding on probing after 1-month re-evaluation alone.

SELECTION OF CITATIONS
SEARCH DETAIL
...