RESUMO
mRNA-based nonviral gene therapy has played an important role in cancer therapy, however, the limited delivery efficiency and therapeutic capacity still require further exploration and enhancement. Immunogene therapy provides a strategy for cancer treatment. Bacteria are tiny single-celled living organisms, many of which can be found in and on the human body and are beneficial to humans. Lactobacillus reuteri is a bacterial member of the gut flora, and recent research has shown that it can reduce intestinal inflammation by stimulating an immunomodulatory response. L. reuteri lysate represents an ideal resource for constructing advanced mRNA delivery systems with immune stimulation potential. Here, we prepared a bifunctional mRNA delivery system DMP-Lac (DOTAP-mPEG-PCL-L. reuteri lysate), which successfully codelivered L. reuteri lysate and IL-23A mRNA, exhibited a high mRNA delivery efficiency of 75.56% ± 0.85%, and strongly promoted the maturation and activation of the immune system in vivo. Both the CT26 abdominal metastasis model and the lung metastasis model also exhibited a good therapeutic effect, and the tumor inhibition rate of DMP-Lac/IL-23A group reached 97.92%. Protein chip technology verified that DMP acted as an immune adjuvant, demonstrating that the L. reuteri lysate could regulate the related immune cells, while IL-23 mRNA caused changes in downstream factors, thus producing the corresponding tumor treatment effect. The DMP-Lac/IL-23A complex exhibited strong anticancer immunotherapeutic effects. Our results demonstrated that this bifunctional mRNA formulation served as a tumor-specific nanomedicine, providing an advanced strategy for colon cancer immunogene therapy.
Assuntos
Neoplasias do Colo , Terapia Genética , Imunoterapia , RNA Mensageiro , Animais , RNA Mensageiro/genética , Neoplasias do Colo/terapia , Neoplasias do Colo/patologia , Camundongos , Terapia Genética/métodos , Humanos , Limosilactobacillus reuteri/química , Camundongos Endogâmicos BALB C , Linhagem Celular Tumoral , Interleucina-23 , Feminino , Lisados BacterianosRESUMO
The probiotic properties of Lactobacillus reuteri (L. reuteri) and its impact on immune function are well-documented. Lipoteichoic acid (LTA) is a crucial immune molecule in Gram-positive bacteria. Despite extensive research on LTA's structural diversity, the immunomodulatory mechanisms of L. reuteri LTA remain largely unexplored. This study investigates the immunomodulatory effects of L. reuteri L1 LTA at various concentrations on RAW 264.7 cells and mice under normal and inflammatory conditions. We found that LTA does not significantly affect healthy subjects; however, low-concentration LTA can reduce inflammation induced by LPS in cells and mice, enhancing the abundance of dominant intestinal bacteria. In contrast, high-concentration LTA exacerbates intestinal damage and dysbiosis. Creatinine may play a role in this differential response. In summary, while LTA does not alter immune homeostasis in healthy organisms, low-concentration LTA may mitigate damage from immune imbalance, but high-concentration LTA can worsen it. This suggests a quantitative requirement for probiotic intake. Our study provides critical theoretical support for understanding the immunomodulatory effects of probiotics on the host and paves the way for future research into the immune mechanisms of probiotics.
Assuntos
Limosilactobacillus reuteri , Lipopolissacarídeos , Probióticos , Ácidos Teicoicos , Ácidos Teicoicos/farmacologia , Animais , Camundongos , Limosilactobacillus reuteri/imunologia , Limosilactobacillus reuteri/química , Lipopolissacarídeos/farmacologia , Células RAW 264.7 , Probióticos/farmacologia , Probióticos/administração & dosagem , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/química , Humanos , Masculino , Fatores Imunológicos/farmacologia , Camundongos Endogâmicos C57BL , FemininoRESUMO
INTRODUCTION: Cystic fibrosis (CF) patients frequently experience gut microbiota dysbiosis. Probiotic supplementation is a potential therapeutic approach to modify gut microbiota and improve CF management through the gut-lung axis. The aim of this study was to investigate the effect of Lactobacillus reuteri supplementation on pulmonary function test, respiratory symptoms and growth in CF patients. METHODS: A randomized, placebo-controlled clinical trial was carried out on 40 children with CF aged from 6 to 20 years. Participants were designated to receive either L. reuteri or placebo daily for 4 months. Pulmonary function tests, weight, height and body mass index (BMI) z-scores were measured pre and post treatment. RESULTS: The median baseline BMI of the patients was 16.28 kg m-2. A significant change in the probiotic group's BMI z-score after the study period was observed (P = 0.034) but not for weight and height z-scores (P > 0.05). After treatment, Pseudomonas aeruginosa grew in sputum cultures of seven in the placebo and one patient in the intervention group (P = 0.03) while at baseline it grew in the sputum of four patients in each group. There was no significant difference in forced expiratory volume in the first second, forced expiratory flow at 25-75% or forced vital capacity change between the two groups after the treatment period (P > 0.05). Additionally, no significant differences were found in pulmonary exacerbations, hospitalization frequencies or COVID-19 infection between the two groups during the study (P > 0.05). CONCLUSION: The results suggest that L. reuteri supplementation may impact the growth of severely malnourished CF patients. Furthermore, it may be concluded that this strain might reduce P. aeruginosa in the sputum culture of CF patients. © 2024 Society of Chemical Industry.
Assuntos
Fibrose Cística , Limosilactobacillus reuteri , Pulmão , Probióticos , Testes de Função Respiratória , Humanos , Fibrose Cística/microbiologia , Fibrose Cística/fisiopatologia , Fibrose Cística/terapia , Masculino , Probióticos/administração & dosagem , Feminino , Adolescente , Criança , Adulto Jovem , Pulmão/microbiologia , Pulmão/fisiopatologia , Adulto , Pseudomonas aeruginosa , Índice de Massa CorporalRESUMO
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.
Assuntos
Limosilactobacillus reuteri , Macrófagos , Mitocôndrias , Cicatrização , Macrófagos/imunologia , Macrófagos/metabolismo , Cicatrização/imunologia , Cicatrização/fisiologia , Animais , Camundongos , Mitocôndrias/metabolismo , Humanos , Modelos Animais de Doenças , Pele/microbiologia , Pele/imunologia , Pele/metabolismoRESUMO
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.
Assuntos
Modelos Animais de Doenças , Limosilactobacillus reuteri , Camundongos Endogâmicos C57BL , Probióticos , Ácido gama-Aminobutírico , Animais , Limosilactobacillus reuteri/metabolismo , Camundongos , Ácido gama-Aminobutírico/metabolismo , Probióticos/administração & dosagem , Probióticos/uso terapêutico , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Macrófagos/metabolismo , Microbioma Gastrointestinal , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevenção & controleRESUMO
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.
Assuntos
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Traumatismo por Reperfusão , Camundongos , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/uso terapêutico , Palmitoilcarnitina/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/tratamento farmacológico , IsquemiaRESUMO
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.
Assuntos
Microbioma Gastrointestinal , Melatonina , Animais , Camundongos , Escherichia coli , Fator 88 de Diferenciação Mieloide/genética , Proteínas Adaptadoras de Transdução de Sinal , Células EpiteliaisRESUMO
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.
Assuntos
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Neuroblastoma , Ratos , Humanos , Camundongos , Animais , Interleucina-6/genética , Depressão/terapia , Fator de Necrose Tumoral alfa/genética , Lipopolissacarídeos/toxicidade , Corticosterona/farmacologia , Serotonina/farmacologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Ansiedade/terapia , Ansiedade/etiologia , Camundongos Endogâmicos C57BLRESUMO
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.
Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus casei , Limosilactobacillus reuteri , Neoplasias Pancreáticas , Camundongos , Humanos , Animais , Receptor 4 Toll-Like/metabolismo , Camundongos Nus , Cromatografia Líquida , Oxipurinol/metabolismo , Oxipurinol/farmacologia , Espectrometria de Massas em Tandem , Macrófagos/metabolismo , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Neoplasias PancreáticasRESUMO
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.
Assuntos
Cólica , Neoplasias Colorretais , Doenças do Sistema Digestório , Diverticulite , Infecções por Helicobacter , Helicobacter pylori , Doenças Inflamatórias Intestinais , Limosilactobacillus reuteri , Humanos , Infecções por Helicobacter/terapia , Doenças do Sistema Digestório/terapia , Constipação Intestinal , Dor Abdominal , DiarreiaRESUMO
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.
Assuntos
Limosilactobacillus reuteri , Osteoporose Pós-Menopausa , Osteoporose , Feminino , Animais , Ratos , Humanos , Ratos Wistar , Fluoreto de Cálcio , Fosfatase Alcalina , Cálcio , Osteoporose/tratamento farmacológico , Estrogênios , Suplementos NutricionaisRESUMO
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.
RESUMO
BACKGROUND: Breast cancer is the most fatal type of cancer in women worldwide. Many chemotherapeutics targeted breast cancer however, they have frightening side effects. One method of controlling cancer cell growth is targeting apoptosis. OBJECTIVE: This study aimed to induce apoptosis in breast cancer cells by purifying L-asparaginase from human breast milk Lactobacillus reuteri isolates via inhibition of Caspases 8 and 9. METHODS: The best L. reuteri isolates producing L-asparagine with the highest enzyme activity were identified from human breast milk and chosen for L-asparaginase purification. The MTT cell viability assay used for measure the toxicity of the enzyme. Breast cancer cell line was used to study the effect of the enzyme on the caspase 8 and caspase 9 gene expression. RESULTS: The MTT cell viability assay showed the inhibition rates ranged between 30% and 80%, of cell death, occurred when 3.125, 6.25, 12.5, 25, 50, and 100 µg/ml of the enzyme used and IC50 was 4.305 µg/ml. The breast cell lines were treated with the enzyme at a concentration of IC50 value. The Cas8 and Cas9 genes expression in L-asparagine treated breast cancer cell line at a concentration of IC50 value were upregulated (the fold of gene expression are 2.071 and 1.197 respectively). CONCLUSIONS: Breast milk L. reuteri L-asparaginase induces apoptosis via Cas8 and Cas9 upregulation in the breast cancer cell line. L. reuteri L-asparaginase treatment may be the hopeful approach for the management of breast cancer. Furthermore, the results may highlight the fact that the presence of L-asparaginase-producing L. reuteri isolates in human breast milk may aid in breast cancer improvement or even prevention.
Assuntos
Neoplasias da Mama , Limosilactobacillus reuteri , Humanos , Feminino , Caspases , Asparaginase/farmacologia , Caspase 8/genética , Caspase 9/genética , Asparagina , Neoplasias da Mama/tratamento farmacológico , Leite Humano , Apoptose , Células MCF-7RESUMO
Many synbiotics are effective for the prevention and treatment of type 2 diabetes mellitus (T2DM). In the treatment of T2DM, synbiotics often regulate the composition of intestinal flora, which autoinducer-2 (AI-2) may play an important role. Whether the changes of intestinal flora are related to AI-2 during synbiotics treatment of T2DM is a topic worth studying. We elucidated the effects of synbiotic composed of mangiferin and Lactobacillus reuteri 1-12 (SML) on T2DM rats. Male Spraque-Dawley rats were injected intraperitoneally with streptozotocin (STZ) and randomly grouped. After that, biochemical parameters, intestinal flora, fecal AI-2, and intestinal colonization of L. reuteri were detected. The results showed that SML had a hypoglycemic effect and mitigated the organ lesions of the liver and pancreas. Also, SML regulated biochemical parameters such as short chain fatty acids (SCFAs), lipopolysaccharides (LPS), intercellular cell adhesion molecule-1 (ICAM-1), and tumor necrosis factor-α (TNF-α). On the other hand, the proportion of probiotics, such as Lactobacillus acidophilus, L. reuteri, Bifidobacterium pseudolongum, Lactobacillus murinus, and Lactobacillus johnsonii, were elevated by the treatment of SML. In addition, SML promoted the colonization and proliferation of L. reuteri in the gut. Another thing to consider was that AI-2 was positively correlated with the total number of OTUs sequences and SML boosted AI-2 in the gut. Taken together, these results supported that SML may modulate intestinal flora through AI-2 to treat T2DM. This study provided a novel alternative strategy for the treatment of T2DM in future.
RESUMO
In our previous studies, Lactobacillus reuteri B1/1, which was renamed Limosilactobacillus reuteri (L. reuteri), was able to modulate the production of pro-inflammatory cytokines and other components of the innate immune response in vitro and in vivo. In this study, we evaluated the effect of Lactobacillus reuteri B1/1 in two concentrations (1 × 107 and 1 × 109 CFU) on the metabolic activity, adherence ability and relative gene expression of pro-inflammatory interleukins (IL-1ß, IL-6, IL-8, IL-18), lumican and olfactomedin 4 produced by non-carcinogenic porcine-derived enterocytes (CLAB). CLAB cells were cultured in a 12-well cell culture plate at a concentration of 4 × 105 cells/well in DMEM medium in a controlled humidified atmosphere for 48 h. A 1 mL volume of each probiotic bacterial suspension was added to the CLAB cells. Plates were incubated for 2 h and 4 h. Our results revealed that L. reuteri B1/1 was able to adhere to CLAB cells in sufficient numbers in both concentrations. In particular, the concentration of 109L. reuteri B1/1 allowed to modulate the gene expression of pro-inflammatory cytokines, as well as to increase the metabolic activity of the cells. In addition, administration of L. reuteri B1/1 in both concentrations significantly stimulated gene expression for both proteins in the CLAB cell line after 4 h of incubation.
RESUMO
Stem cell injection is good for periodontal regeneration due to the capacity of stem cells to differentiate toward osteogenic direction and to regulate the production of pro- and anti-inflammatory cytokines. However, injected cells are difficult to track in vivo. And there is microbiota in oral cavity, the dysbiosis of which leads to the damage and loss of periodontal tissue. Here, we demonstrated an enhanced periodontal repair was due to an altered oral microbiota. Periodontal defects were surgically prepared in rats, and periodontal ligament stem cells (PDLSCs) labeled by superparamagnetic iron oxide (SPIO) nanoparticles (PC-SPIO) were injected, with PDLSCs and saline treatments as controls. Detected by magnetic resonance imaging (MRI) and histological staining, PC-SPIO was major at limited areas in regenerated periodontal tissues. PC-SPIO-treated rats achieved better periodontal regeneration than the other two groups. Concurrently, the oral microbiota of PC-SPIO-treated rats was changed, presenting SPIO-Lac as a biomarker. SPIO-Lac assisted periodontal repair in vivo, inhibited the inflammation of macrophages induced by lipopolysaccharide (LPS) and antibacterial in vitro. Therefore, our study proved that SPIO-labeled cells can be tracked in periodontal defect and highlighted a potential positive role of an oral microbiota in periodontal regeneration, suggesting the possibility of periodontal repair promotion by manipulating oral microbiota.
RESUMO
The use of probiotics by cancer patients is increasing, including among those undergoing immune checkpoint inhibitor (ICI) treatment. Here, we elucidate a critical microbial-host crosstalk between probiotic-released aryl hydrocarbon receptor (AhR) agonist indole-3-aldehyde (I3A) and CD8 T cells within the tumor microenvironment that potently enhances antitumor immunity and facilitates ICI in preclinical melanoma. Our study reveals that probiotic Lactobacillus reuteri (Lr) translocates to, colonizes, and persists within melanoma, where via its released dietary tryptophan catabolite I3A, it locally promotes interferon-γ-producing CD8 T cells, thereby bolstering ICI. Moreover, Lr-secreted I3A was both necessary and sufficient to drive antitumor immunity, and loss of AhR signaling within CD8 T cells abrogated Lr's antitumor effects. Further, a tryptophan-enriched diet potentiated both Lr- and ICI-induced antitumor immunity, dependent on CD8 T cell AhR signaling. Finally, we provide evidence for a potential role of I3A in promoting ICI efficacy and survival in advanced melanoma patients.
Assuntos
Limosilactobacillus reuteri , Melanoma , Microambiente Tumoral , Humanos , Dieta , Inibidores de Checkpoint Imunológico , Limosilactobacillus reuteri/metabolismo , Melanoma/terapia , Triptofano/metabolismo , Linfócitos T CD8-Positivos/imunologia , Receptores de Hidrocarboneto Arílico/agonistasRESUMO
Objective: Gut microbiota play a key role in the pathogenesis of obesity and depression. Probiotics are a preventive strategy for obesity and a novel treatment for depression symptoms. However, the ameliorative or therapeutic effect of potential probiotic candidate Lactobacillus reuteri (L. reuteri) on obesity and depression comorbidity still remains unclear. We investigated the effects of chronic unpredictable mild stress (CUMS) in high-fat diet-fed mice and the effects of Lactobacillus reuteri strain 8008 on various disease indicators of obesity and depression comorbidity disease. Methods: Forty male C57BL/6 mice were randomized into 2 groups: the normal control (NC) group (n = 10) and the high-fat diet (HFD) group (n = 30), being fed with normal diet (ND) or high-fat diet (HFD) for 8 weeks, respectively. Then the obese mice fed with HFD were randomly allocated into 3 sub-groups: the HFD group (n = 10); the HFD + CUMS group (n = 10); the HFD + CUMS + L.r group (n = 10). The latter 2 subgroups underwent CUMS for 4 weeks to build the obesity and depression comorbidity mice model. During the duration of treatment, mice were gavaged with 0.5 mL PBS solution or L. reuteri (2 × 109 CFU/mL) once a day, respectively. The body weight, food intake, organ weight, behavioral indicators, histology, blood lipids, levels of inflammatory cytokines and tight junction proteins and abundance of colonic contents bacteria were measured. Results: The obesity and depression comorbidity mice model was successfully established after HFD feeding and chronic stress. The comorbid mice demonstrated inflammatory responses increase in liver and adipose tissues, worsened damage to the intestinal barrier as well as gut microbiota disorder. Gavaged with L. reuteri attenuated depressive-like behavior, improved blood lipids and insulin resistance, reduced inflammation in liver and adipose tissues, improved intestinal tight junctions as well as the microbiome dysbiosis in obesity and depression comorbidity mice. Conclusion: Lactobacillus reuteri strain 8008 could alleviate depressive-like behaviors and related indicators of obesity disorders by regulating the gut microbiota in obesity and depression comorbid mice.
RESUMO
Lactobacillus reuteri is a probiotic with bacteriostatic effects, which can effectively inhibit the activity of pathogens. However, the molecular mechanism underlying the inhibition of pathogens by L. reuteri in intestinal cells remains unclear. Using the porcine intestinal cell line IPEC-J2 as a model, we combined RNA-seq and ATAC-seq methods to delineate the porcine genome-wide changes in biological processes and chromatin accessibility in IPEC-J2 cells stimulated by Salmonella enterica BNCC186354, as well as L. reuteri ATCC 53608. Overall, we found that many porcine transcripts were altered after S. enterica BNCC186354 treatment, while L. reuteri ATCC 53608 treatment partially restored this alteration, such as salmonella infection and PI3K/AKT and MAPK pathways. Combined analysis of these two datasets revealed that 26 genes with similar trends overlapped between gene expression and chromatin accessibility. In addition, we identified potential host functional transcription factors (TFs), such as GATA1, TAL1, TBP, RUNX1, Gmeb1, Gfi1b, RARA, and RXRG, in IPEC-J2 cells that might play a critical role and are targeted by L. reuteri ATCC 53608. Moreover, we verified that PI3K/AKT, MAPK, and apoptosis pathways are potentially regulated by S. enterica BNCC186354 but restored by L. reuteri ATCC 53608. The PI3K/AKT pathway was activated by L. reuteri ATCC 53608, thereby potentially inhibiting S. enterica BNCC186354 infection. In conclusion, our data provide new insights into the expression pattern of functional genes and the epigenetic alterations in IPEC-J2 cells underlying the bacteriostatic action of L. reuteri ATCC 53608.
Assuntos
Limosilactobacillus reuteri , Salmonella enterica , Animais , Suínos , Cromatina , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-aktRESUMO
Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Thus, strategies to mitigate toxicity are important and can revitalize the current standard of care. We previously established that LR-IL-22 protects the intestine from WAI. We now hypothesize that LR-IFN-ß is an effective radiation protector and mitigator and is rapidly cleared from the digestive tract, making it an option for intestinal radioprotection. We report that the gavage of LR-IFN-ß during WAI provides improved intestinal barrier integrity and significantly preserves the numbers of Lgr5+GFP+ intestinal stem cells, improving survival. The rapid clearance of the genetically engineered probiotic from the digestive tract renders it a safe and feasible radiation mitigator. Therefore, the above genetically engineered probiotic is both a feasible and effective radiation mitigator that could potentially revolutionize the management of OC patients. Furthermore, the subsequent addition of platinum/taxane-based chemotherapy to the combination of WAI and LR-IFN-ß should reduce tumor volume while protecting the intestine and should improve the overall survival in OC patients.