Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.399
1.
Theranostics ; 14(7): 2719-2735, 2024.
Article En | MEDLINE | ID: mdl-38773969

Aim: To elucidate dynamics and functions in colonic macrophage subsets, and their regulation by Bifidobacterium breve (B. breve) and its associated metabolites in the initiation of colitis-associated colorectal cancer (CAC). Methods: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to create a CAC model. The tumor-suppressive effect of B. breve and variations of macrophage subsets were evaluated. Intestinal macrophages were ablated to determine their role in the protective effects of B. breve. Efficacious molecules produced by B. breve were identified by non-targeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The molecular mechanism was further verified in murine bone marrow-derived macrophages (BMDMs), macrophages derived from human peripheral blood mononuclear cells (hPBMCs), and demonstrated in CAC mice. Results: B. breve alleviated colitis symptoms, delayed colonic tumorigenesis, and promoted phenotypic differentiation of immature inflammatory macrophages into mature homeostatic macrophages. On the contrary, the ablation of intestinal macrophages largely annulled the protective effects of B. breve. Microbial analysis of colonic contents revealed the enrichment of probiotics and the depletion of potential pathogens following B. breve supplementation. Moreover, indole-3-lactic acid (ILA) was positively correlated with B. breve in CAC mice and highly enriched in the culture supernatant of B. breve. Also, the addition of ILA directly promoted AKT phosphorylation and restricted the pro-inflammatory response of murine BMDMs and macrophages derived from hPBMCs in vitro. The effects of ILA in murine BMDMs and macrophages derived from hPBMCs were abolished by the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the AKT inhibitor MK-2206. Furthermore, ILA could protect against tumorigenesis by regulating macrophage differentiation in CAC mice; the AhR antagonist largely abrogated the effects of B. breve and ILA in relieving colitis and tumorigenesis. Conclusion: B. breve-mediated tryptophan metabolism ameliorates the precancerous inflammatory intestinal milieu to inhibit tumorigenesis by directing the differentiation of immature colonic macrophages.


Bifidobacterium breve , Cell Differentiation , Colitis , Indoles , Macrophages , Probiotics , Animals , Mice , Macrophages/metabolism , Macrophages/drug effects , Bifidobacterium breve/metabolism , Indoles/pharmacology , Indoles/metabolism , Humans , Colitis/chemically induced , Colitis/microbiology , Colitis/complications , Cell Differentiation/drug effects , Probiotics/pharmacology , Probiotics/administration & dosage , Disease Models, Animal , Carcinogenesis/drug effects , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/metabolism , Mice, Inbred C57BL , Colon/microbiology , Colon/pathology , Colon/metabolism , Dextran Sulfate , Male , Gastrointestinal Microbiome , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Azoxymethane
2.
Front Immunol ; 15: 1394501, 2024.
Article En | MEDLINE | ID: mdl-38774883

Extracellular vesicles (EVs) are cell-derived membrane-surrounded vesicles that carry bioactive molecules. Among EVs, outer membrane vesicles (OMVs), specifically produced by Gram-negative bacteria, have been extensively characterized and their potential as vaccines, adjuvants or immunotherapeutic agents, broadly explored in mammals. Nonetheless, Gram-positive bacteria can also produce bilayered spherical structures from 20 to 400 nm involved in pathogenesis, antibiotic resistance, nutrient uptake and nucleic acid transfer. However, information regarding their immunomodulatory potential is very scarce, both in mammals and fish. In the current study, we have produced EVs from the Gram-positive probiotic Bacillus subtilis and evaluated their immunomodulatory capacities using a rainbow trout intestinal epithelial cell line (RTgutGC) and splenic leukocytes. B. subtilis EVs significantly up-regulated the transcription of several pro-inflammatory and antimicrobial genes in both RTgutGC cells and splenocytes, while also up-regulating many genes associated with B cell differentiation in the later. In concordance, B. subtilis EVs increased the number of IgM-secreting cells in splenocyte cultures, while at the same time increased the MHC II surface levels and antigen-processing capacities of splenic IgM+ B cells. Interestingly, some of these experiments were repeated comparing the effects of B. subtilis EVs to EVs obtained from another Bacillus species, Bacillus megaterium, identifying important differences. The data presented provides evidence of the immunomodulatory capacities of Gram-positive EVs, pointing to the potential of B. subtilis EVs as adjuvants or immunostimulants for aquaculture.


Bacillus subtilis , Extracellular Vesicles , Leukocytes , Oncorhynchus mykiss , Spleen , Animals , Bacillus subtilis/immunology , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Oncorhynchus mykiss/immunology , Oncorhynchus mykiss/microbiology , Spleen/immunology , Spleen/cytology , Leukocytes/immunology , Leukocytes/metabolism , Probiotics/pharmacology , Cell Line , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Immunomodulation , Intestines/immunology
3.
Sci Rep ; 14(1): 11560, 2024 May 21.
Article En | MEDLINE | ID: mdl-38773299

IBD is a disorder which could be caused by oxidative stress. This investigation aims to determine if probiotics and postbiotics can control oxidative stress and inflammation and compare the effectiveness of these two probiotic and postbiotic mixtures of substances. 88 strains of Lactobacillus and Bifidobacterium were tested for antioxidant activity. Male wild-type C57BL/6 mice were divided into four experimental groups, namely high fat diet (HFD) + PBS, HFD + DSS, HFD + DSS + 109 cfu/ml of probiotics, and HFD + DSS + 109 cfu/ml of postbiotics. The phenotypical indices and pathological scores were assessed. The expression of genes related to NF-kB and Nrf2 signaling pathways and enzymes associated with oxidant/anti-oxidant activities, and proinflammatory/inflammatory cytokines were assessed. In contrast to the groups exposed to DSS, mice treated with probiotics mixture and postbiotics mixture alongside DSS displayed alleviation of DSS-induced adverse effects on phenotypical characteristics, as well as molecular indices such as the Nrf2 and NF-kB related genes, with a greater emphasis on the postbiotics component. In accordance with the findings of the present investigation, it can be inferred that even in using a high-fat dietary regimen as an inducer of oxidative stress, the emergence of inflammation can be effectively addressed through the utilization of probiotics and, more specifically, postbiotics.


Anti-Inflammatory Agents , Antioxidants , Colitis , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Probiotics , Signal Transduction , Animals , Probiotics/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Male , Mice , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Anti-Inflammatory Agents/pharmacology , Signal Transduction/drug effects , Oxidative Stress/drug effects , Lactobacillus , Bifidobacterium , Diet, High-Fat/adverse effects
4.
Nutrients ; 16(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732619

Functional foods with probiotics are safe and effective dietary supplements to improve overweight and obesity. Thus, altering the intestinal microflora may be an effective approach for controlling or preventing obesity. This review aims to summarize the experimental method used to study probiotics and obesity, and recent advances in probiotics against obesity. In particular, we focused on studies (in vitro and in vivo) that used probiotics to treat obesity and its associated comorbidities. Several in vitro and in vivo (animal and human clinical) studies conducted with different bacterial species/strains have reported that probiotics promote anti-obesity effects by suppressing the differentiation of pre-adipocytes through immune cell activation, maintaining the Th1/Th2 cytokine balance, altering the intestinal microbiota composition, reducing the lipid profile, and regulating energy metabolism. Most studies on probiotics and obesity have shown that probiotics are responsible for a notable reduction in weight gain and body mass index. It also increases the levels of anti-inflammatory adipokines and decreases those of pro-inflammatory adipokines in the blood, which are responsible for the regulation of glucose and fatty acid breakdown. Furthermore, probiotics effectively increase insulin sensitivity and decrease systemic inflammation. Taken together, the intestinal microbiota profile found in overweight individuals can be modified by probiotic supplementation which can create a promising environment for weight loss along enhancing levels of adiponectin and decreasing leptin, tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemotactic protein (MCP)-1, and transforming growth factor (TGF)-ß on human health.


Adipogenesis , Anti-Inflammatory Agents , Gastrointestinal Microbiome , Obesity , Probiotics , Probiotics/pharmacology , Probiotics/therapeutic use , Humans , Obesity/microbiology , Animals , Anti-Inflammatory Agents/pharmacology , Inflammation , Adipokines/blood
5.
Mol Biol Rep ; 51(1): 613, 2024 May 05.
Article En | MEDLINE | ID: mdl-38704764

BACKGROUND: The non-alcoholic fatty liver disease (NAFLD) is prevalent in as many as 25% of adults who are afflicted with metabolic syndrome. Oxidative stress plays a significant role in the pathophysiology of hepatic and renal injury associated with NAFLD. Therefore, probiotics such as Lactobacillus casei (LBC) and the microalga Chlorella vulgaris (CV) may be beneficial in alleviating kidney injury related to NAFLD. MATERIALS AND METHODS: This animal study utilized 30 C57BL/6 mice, which were evenly distributed into five groups: the control group, the NAFLD group, the NAFLD + CV group, the NAFLD + LBC group, and the NAFLD + CV + LBC group. A high-fat diet (HFD) was administered to induce NAFLD for six weeks. The treatments with CV and LBC were continued for an additional 35 days. Biochemical parameters, total antioxidant capacity (TAC), and the expression of kidney damage marker genes (KIM 1 and NGAL) in serum and kidney tissue were determined, respectively. A stereological analysis was conducted to observe the structural changes in kidney tissues. RESULTS: A liver histopathological examination confirmed the successful induction of NAFLD. Biochemical investigations revealed that the NAFLD group exhibited increased ALT and AST levels, significantly reduced in the therapy groups (p < 0.001). The gene expression levels of KIM-1 and NGAL were elevated in NAFLD but were significantly reduced by CV and LBC therapies (p < 0.001). Stereological examinations revealed reduced kidney size, volume, and tissue composition in the NAFLD group, with significant improvements observed in the treated groups (p < 0.001). CONCLUSION: This study highlights the potential therapeutic efficacy of C. vulgaris and L. casei in mitigating kidney damage caused by NAFLD. These findings provide valuable insights for developing novel treatment approaches for managing NAFLD and its associated complications.


Chlorella vulgaris , Diet, High-Fat , Kidney , Lacticaseibacillus casei , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Probiotics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/pathology , Animals , Diet, High-Fat/adverse effects , Mice , Kidney/pathology , Kidney/metabolism , Probiotics/pharmacology , Probiotics/administration & dosage , Male , Oxidative Stress/drug effects , Disease Models, Animal , Liver/pathology , Liver/metabolism , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Diseases/therapy , Antioxidants/metabolism
6.
J Nanobiotechnology ; 22(1): 241, 2024 May 12.
Article En | MEDLINE | ID: mdl-38735933

BACKGROUND: Colorectal cancer (CRC) incidence is increasing in recent years due to intestinal flora imbalance, making oral probiotics a hotspot for research. However, numerous studies related to intestinal flora regulation ignore its internal mechanisms without in-depth research. RESULTS: Here, we developed a probiotic microgel delivery system (L.r@(SA-CS)2) through the layer-by-layer encapsulation technology of alginate (SA) and chitosan (CS) to improve gut microbiota dysbiosis and enhance anti-tumor therapeutic effect. Short chain fatty acids (SCFAs) produced by L.r have direct anti-tumor effects. Additionally, it reduces harmful bacteria such as Proteobacteria and Fusobacteriota, and through bacteria mutualophy increases beneficial bacteria such as Bacteroidota and Firmicutes which produce butyric acid. By binding to the G protein-coupled receptor 109A (GPR109A) on the surface of colonic epithelial cells, butyric acid can induce apoptosis in abnormal cells. Due to the low expression of GPR109A in colon cancer cells, MK-6892 (MK) can be used to stimulate GPR109A. With increased production of butyrate, activated GPR109A is able to bind more butyrate, which further promotes apoptosis of cancer cells and triggers an antitumor response. CONCLUSION: It appears that the oral administration of L.r@(SA-CS)2 microgels may provide a treatment option for CRC by modifying the gut microbiota.


Fatty Acids, Volatile , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Gastrointestinal Microbiome/drug effects , Probiotics/pharmacology , Humans , Fatty Acids, Volatile/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Mice , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Administration, Oral , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Receptors, G-Protein-Coupled/metabolism , Microgels/chemistry , Mice, Inbred BALB C , Butyric Acid/pharmacology , Butyric Acid/metabolism
7.
Gut Microbes ; 16(1): 2347722, 2024.
Article En | MEDLINE | ID: mdl-38706205

The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5+ intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.


Gastrointestinal Microbiome , Indoles , Mice, Inbred C57BL , Probiotics , Receptors, Aryl Hydrocarbon , Wnt Signaling Pathway , Animals , Mice , Gastrointestinal Microbiome/drug effects , Male , Humans , Probiotics/administration & dosage , Probiotics/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Indoles/metabolism , Indoles/pharmacology , Radiation-Protective Agents/pharmacology , Organoids/metabolism , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/radiation effects , Intestines/microbiology , Intestines/radiation effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics
8.
Gut Microbes ; 16(1): 2341717, 2024.
Article En | MEDLINE | ID: mdl-38717360

The occurrence and progression of tumors are often accompanied by disruptions in the gut microbiota. Inversely, the impact of the gut microbiota on the initiation and progression of cancer is becoming increasingly evident, influencing the tumor microenvironment (TME) for both local and distant tumors. Moreover, it is even suggested to play a significant role in the process of tumor immunotherapy, contributing to high specificity in therapeutic outcomes and long-term effectiveness across various cancer types. Probiotics, with their generally positive influence on the gut microbiota, may serve as effective agents in synergizing cancer immunotherapy. They play a crucial role in activating the immune system to inhibit tumor growth. In summary, this comprehensive review aims to provide valuable insights into the dynamic interactions between probiotics, gut microbiota, and cancer. Furthermore, we highlight recent advances and mechanisms in using probiotics to improve the effectiveness of cancer immunotherapy. By understanding these complex relationships, we may unlock innovative approaches for cancer diagnosis and treatment while optimizing the effects of immunotherapy.


Gastrointestinal Microbiome , Immunotherapy , Neoplasms , Probiotics , Tumor Microenvironment , Probiotics/therapeutic use , Probiotics/administration & dosage , Probiotics/pharmacology , Humans , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/microbiology , Tumor Microenvironment/immunology , Animals
9.
J Nanobiotechnology ; 22(1): 267, 2024 May 19.
Article En | MEDLINE | ID: mdl-38764014

Enhancing immune response activation through the synergy of effective antigen delivery and immune enhancement using natural, biodegradable materials with immune-adjuvant capabilities is challenging. Here, we present NAPSL.p that can activate the Toll-like receptor 4 (TLR4) pathway, an amphiphilic exopolysaccharide, as a potential self-assembly adjuvant delivery platform. Its molecular structure and unique properties exhibited remarkable self-assembly, forming a homogeneous nanovaccine with ovalbumin (OVA) as the model antigen. When used as an adjuvant, NAPSL.p significantly increased OVA uptake by dendritic cells. In vivo imaging revealed prolonged pharmacokinetics of NAPSL. p-delivered OVA compared to OVA alone. Notably, NAPSL.p induced elevated levels of specific serum IgG and isotype titers, enhancing rejection of B16-OVA melanoma xenografts in vaccinated mice. Additionally, NAPSL.p formulation improved therapeutic effects, inhibiting tumor growth, and increasing animal survival rates. The nanovaccine elicited CD4+ and CD8+ T cell-based immune responses, demonstrating the potential for melanoma prevention. Furthermore, NAPSL.p-based vaccination showed stronger protective effects against influenza compared to Al (OH)3 adjuvant. Our findings suggest NAPSL.p as a promising, natural self-adjuvanting delivery platform to enhance vaccine design across applications.


Adjuvants, Immunologic , Melanoma, Experimental , Mice, Inbred C57BL , Ovalbumin , Probiotics , Animals , Ovalbumin/immunology , Ovalbumin/chemistry , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Probiotics/pharmacology , Melanoma, Experimental/immunology , Female , Dendritic Cells/immunology , Toll-Like Receptor 4/metabolism , Cancer Vaccines/immunology , Cancer Vaccines/chemistry , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Nanoparticles/chemistry , CD4-Positive T-Lymphocytes/immunology
10.
ACS Nano ; 18(20): 12917-12932, 2024 May 21.
Article En | MEDLINE | ID: mdl-38720520

Inflammatory bowel diseases (IBDs) refer to multifaceted disorders in the intestinal microenvironment and microbiota homeostasis. In view of the broad bioactivity and high compatibility of polyphenols, there is considerable interest in developing a polyphenol-based collaborative platform to remodel the IBD microenvironment and regulate microbiota. Here, we demonstrated the coordination assembly of nanostructured polyphenols to modify probiotics and simultaneously deliver drugs for IBD treatment. Inspired by the distinctive structure of tannic acid (TA), we fabricated nanostructured pBDT-TA by using a self-polymerizable aromatic dithiol (BDT) and TA, which exhibited excellent antioxidant and anti-inflammatory capability in vitro. We thus coated pBDT-TA and sodium alginate (SA) to the surface of Escherichia coli Nissle 1917 layer by layer to construct the collaborative platform EcN@SA-pBDT-TA. The modified probiotics showed improved resistance to oxidative and inflammatory stress, which resulted in superior colon accumulation and retention in IBD model mice. Further, EcN@SA-pBDT-TA could alleviate dextran sulfate sodium (DSS)-induced colitis by controlling the inflammatory response, repairing intestinal barriers, and modulating gut microbiota. Importantly, EcN@SA-pBDT-TA-mediated IBD drug delivery could achieve an improved therapeutic effect in DSS model mice. Given the availability and functionality of polyphenol and prebiotics, we expected that nanostructured polyphenol-modified probiotics provided a solution to develop a collaborative platform for IBD treatment.


Inflammatory Bowel Diseases , Nanoparticles , Polyphenols , Probiotics , Tannins , Animals , Probiotics/pharmacology , Probiotics/chemistry , Probiotics/administration & dosage , Polyphenols/chemistry , Polyphenols/pharmacology , Mice , Nanoparticles/chemistry , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/therapy , Tannins/chemistry , Tannins/pharmacology , Mice, Inbred C57BL , Escherichia coli/drug effects , Dextran Sulfate/chemistry , Alginates/chemistry , Alginates/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology
11.
World J Microbiol Biotechnol ; 40(7): 198, 2024 May 10.
Article En | MEDLINE | ID: mdl-38727952

Atherosclerosis is viewed as not just as a problem of lipid build-up in blood vessels, but also as a chronic inflammatory disease involving both innate and acquired immunity. In atherosclerosis, the inflammation of the arterial walls is the key characteristic that significantly contributes to both the instability of plaque and the occlusion of arteries by blood clots. These events ultimately lead to stroke and acute coronary syndrome. Probiotics are living microorganisms that, when consumed in the right quantities, offer advantages for one's health. The primary objective of this study was to investigate the influence of Lactiplantibacillus plantarum ATCC 14917 (ATCC 14917) on the development of atherosclerotic plaques and its underlying mechanism in Apo lipoprotein E-knockout (Apoe-/- mice). In this study, Apoe-/- mice at approximately 8 weeks of age were randomly assigned to three groups: a Normal group that received a normal chow diet, a high fat diet group that received a gavage of PBS, and a Lactiplantibacillus plantarum ATCC 14917 group that received a high fat diet and a gavage of 0.2 ml ATCC 14917 (2 × 109 CFU/mL) per day for a duration of 12 weeks. Our strain effectively reduced the size of plaques in Apoe-/- mice by regulating the expression of inflammatory markers, immune cell markers, chemokines/chemokine receptors, and tight junction proteins (TJPs). Specifically, it decreased the levels of inflammatory markers (ICAM-1, CD-60 MCP-1, F4/80, ICAM-1, and VCAM-1) in the thoracic aorta, (Ccr7, cd11c, cd4, cd80, IL-1ß, TNF-α) in the colon, and increased the activity of ROS-scavenging enzymes (SOD-1 and SOD-2). It also influenced the expression of TJPs (occludin, ZO-1, claudin-3, and MUC-3). In addition, the treatment of ATCC 14917 significantly reduced the level of lipopolysaccharide in the mesenteric adipose tissue. The findings of our study demonstrated that our strain effectively decreased the size of atherosclerotic plaques by modulating inflammation, oxidative stress, intestinal integrity, and intestinal immunity.


Apolipoproteins E , Atherosclerosis , Plaque, Atherosclerotic , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Mice , Atherosclerosis/microbiology , Apolipoproteins E/genetics , Male , Disease Models, Animal , Mice, Knockout , Diet, High-Fat , Lactobacillus plantarum , Cytokines/metabolism , Mice, Inbred C57BL , Inflammation
12.
World J Microbiol Biotechnol ; 40(6): 194, 2024 May 07.
Article En | MEDLINE | ID: mdl-38713319

The development and utilization of probiotics have many environmental benefits when they are used to replace antibiotics in animal production. In this study, intestinal lactic acid bacteria were isolated from the intestines of Cherry Valley ducks. Probiotic lactic acid bacterial strains were screened for antibacterial activity and tolerance to produce a Lactobacillus spp. mixture. The effects of the compound on the growth performance and intestinal flora of Cherry Valley ducks were studied. Based on the results of the antibacterial activity and tolerance tests, the highly active strains Lactobacillus casei 1.2435, L. salivarius L621, and L. salivarius L4 from the intestines of Cherry Valley ducks were selected. The optimum ratio of L. casei 1.2435, L. salivarius L621, and L. salivarius L4 was 1:1:2, the amount of inoculum used was 1%, and the fermentation time was 14 h. In vivo experiments showed that compared with the control group, the relative abundances of intestinal Lactobacillus and Blautia were significantly increased in the experimental group fed the lactobacilli compound (P < 0.05); the relative abundances of Parabacteroides, [Ruminococcus]_torques_group, and Enterococcus were significantly reduced (P < 0.05), and the growth and development of the dominant intestinal flora were promoted in the Cherry Valley ducks. This study will provide more opportunities for Cherry Valley ducks to choose microecological agents for green and healthy breeding.


Ducks , Gastrointestinal Microbiome , Intestines , Lactobacillus , Probiotics , Animals , Probiotics/pharmacology , Ducks/microbiology , Gastrointestinal Microbiome/drug effects , Lactobacillus/isolation & purification , Intestines/microbiology , Fermentation , Animal Feed , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/pharmacology
13.
Bull Exp Biol Med ; 176(5): 543-547, 2024 Mar.
Article En | MEDLINE | ID: mdl-38717566

We studied the dynamics of the main hemodynamic parameters in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats with visceral obesity and chemically induced colitis (CIC) against the background of probiotic therapy. Systolic BP, HR, and body temperature were recorded over 36 days using a wireless telemetry system. During 8 days (3 days before CIC induction and until the end of the experiment) the animals were intragastrically administered a probiotic based on Lactobacillus delbrueckii D5 strain. At baseline, systolic BP was significantly higher in the SHR group, while HR and body temperature did not differ in SHR and WKY rats. On day 8 after CIC induction, systolic BP, HR, and body temperature in SHR were significantly increased in comparison with the initial values. In the group of WKY rats, all indices at the end of the experiment remained at the initial levels. Probiotic therapy in SHR, in contrast to WKY rats, did not lead to normalization of body temperature and hemodynamic disorders resulting from CIC.


Body Temperature , Colitis , Hemodynamics , Probiotics , Rats, Inbred SHR , Rats, Inbred WKY , Animals , Probiotics/pharmacology , Probiotics/administration & dosage , Rats , Male , Colitis/chemically induced , Colitis/physiopathology , Colitis/microbiology , Hemodynamics/drug effects , Body Temperature/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Heart Rate/drug effects , Lactobacillus delbrueckii , Obesity/physiopathology , Obesity, Abdominal/physiopathology , Obesity, Abdominal/chemically induced
14.
Fish Shellfish Immunol ; 149: 109618, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729251

An eight-week feeding trial was designed to assess which component of commensal Bacillus siamensis LF4 can mitigate SBM-induced enteritis and microbiota dysbiosis in spotted seabass (Lateolabrax maculatus) based on TLRs-MAPKs/NF-кB signaling pathways. Fish continuously fed low SBM (containing 16 % SBM) and high SBM (containing 40 % SBM) diets were used as positive (FM group) and negative (SBM group) control, respectively. After feeding high SBM diet for 28 days, fish were supplemented with B. siamensis LF4-derived whole cell wall (CW), cell wall protein (CWP), lipoteichoic acid (LTA) or peptidoglycan (PGN) until 56 days. The results showed that a high inclusion of SBM in the diet caused enteritis, characterized with significantly (P < 0.05) decreased muscular thickness, villus height, villus width, atrophied and loosely arranged microvillus. Moreover, high SBM inclusion induced an up-regulation of pro-inflammatory cytokines and a down-regulation of occludin, E-cadherin, anti-inflammatory cytokines, apoptosis related genes and antimicrobial peptides. However, dietary supplementation with CW, LTA, and PGN of B. siamensis LF4 could effectively alleviate enteritis caused by a high level of dietary SBM. Additionally, CWP and PGN administration increased beneficial Cetobacterium and decreased pathogenic Plesiomonas and Brevinema, while dietary LTA decreased Plesiomonas and Brevinema, suggesting that CWP, LTA and PGN positively modulated intestinal microbiota in spotted seabass. Furthermore, CW, LTA, and PGN application significantly stimulated TLR2, TLR5 and MyD88 expressions, and inhibited the downstream p38 and NF-κB signaling. Taken together, these results suggest that LTA and PGN from B. siamensis LF4 could alleviate soybean meal-induced enteritis and microbiota dysbiosis in L. maculatus, and p38 MAPK/NF-κB pathways might be involved in those processes.


Animal Feed , Bacillus , Diet , Dysbiosis , Enteritis , Fish Diseases , Gastrointestinal Microbiome , Glycine max , Lipopolysaccharides , Peptidoglycan , Teichoic Acids , Animals , Fish Diseases/immunology , Animal Feed/analysis , Enteritis/veterinary , Enteritis/immunology , Enteritis/microbiology , Dysbiosis/veterinary , Dysbiosis/immunology , Bacillus/physiology , Bacillus/chemistry , Gastrointestinal Microbiome/drug effects , Diet/veterinary , Glycine max/chemistry , Lipopolysaccharides/pharmacology , Teichoic Acids/pharmacology , Peptidoglycan/pharmacology , Peptidoglycan/administration & dosage , Bass/immunology , Probiotics/pharmacology , Probiotics/administration & dosage , Dietary Supplements/analysis , Random Allocation
15.
Food Funct ; 15(10): 5496-5509, 2024 May 20.
Article En | MEDLINE | ID: mdl-38690869

Postbiotics have been proposed as clinically viable alternatives to probiotics, addressing limitations and safety concerns associated with probiotic use. However, direct comparisons between the functional differences and health benefits of probiotics and postbiotics remain scarce. This study compared directly the desensitization effect of probiotics and postbiotics derived from Lactiplantibacillus plantarum strain DPUL-F232 in the whey protein-induced allergic rat model. The results demonstrate that administering both live and heat killed F232 significantly alleviated allergy symptoms, reduced intestinal inflammation, and decreased serum antibody and histamine levels in rats. Both forms of F232 were effective in regulating the Th1/Th2 balance, promoting the secretion of the regulatory cytokine IL-10, inhibiting mast cell degranulation and restoring the integrity of the intestinal barrier through the upregulation of tight junction proteins. Considering the enhanced stability and reduced safety concerns of postbiotics compared to probiotics, alongside their ability to regulate allergic reactions, we suggest that postbiotics may serve as viable substitutes for probiotics in managing food allergies and potentially other diseases.


Food Hypersensitivity , Probiotics , Whey Proteins , Animals , Whey Proteins/pharmacology , Rats , Probiotics/pharmacology , Lactobacillus plantarum , Rats, Sprague-Dawley , Intestinal Mucosa/immunology , Male , Female , Hot Temperature , Humans
16.
Fish Shellfish Immunol ; 149: 109595, 2024 Jun.
Article En | MEDLINE | ID: mdl-38692381

This study aimed to elucidate the effects of dietary fermented products of Bacillus velezensis T23 on the growth, immune response and gut microbiota in Pacific white shrimp (Litopenaeus vannamei). Shrimp were fed with diets containing fermentation products of B. velezensis T23 at levels of (0, 0.05, 0.1, 0.2, 0.3, 0.4, and 0.5 g/kg) for 4 weeks, to assess the influence on shrimp growth. The results showed that 0.3 and 0.4 g/kg T23 supplementation improved shrimp growth and feed utilization. Based on these results we selected these three diets (Control, 0.3T23 and 0.4T23) to assess the effect on immune response and gut microbiota of shrimp. Compared with the control, the 0.3T23 and 0.4T23 groups enhanced lipase and α-amylase activities in the gut significantly. Moreover, the 0.4T23 group decreased TAG and MDA levels in hepatopancreas, ALT and AST levels of serum significantly (P < 0.05). In hepatopancreas, CAT and SOD activities were improved observably and the MDA content was reduced markedly in both T23 groups. The expressions of antimicrobial related genes, Cru and peroxinectin in the 0.3T23 group, and proPO and peroxinectin in the 0.4T23 group were up-regulated remarkably (P < 0.05). Moreover, hepatopancreas of shrimp fed with a diet amended with T23 showed a significant down-regulated expression of nf-kb and tnf-α genes, while expressions of tgf-ß was considerably up-regulated. Furthermore, serum LPS and LBP contents were reduced markedly in T23 groups. Intestinal SOD and CAT were noteworthy higher in T23 groups (P < 0.05). In the intestine of shrimp fed on the diet enriched with T23 the expression of nf-κb and tnf-α exhibited markedly down-regulated, whereas hif1α was up-regulated (P < 0.05). Besides, in the intestine of shrimp grouped under T23, Cru and peroxinectin genes were markedly up-regulated (P < 0.05). Dietary 0.3 g/kg T23 also upregulated the ratio of Rhodobacteraceae to Vibrionaceae in the gut of the shrimp. Taken together, the inclusion of B. velezensis T23 in the diet of shrimp enhanced the growth and feed utilization, enhanced hepatopancreas and intestine health.


Animal Feed , Bacillus , Diet , Hepatopancreas , Intestines , Penaeidae , Probiotics , Animals , Penaeidae/immunology , Penaeidae/growth & development , Penaeidae/microbiology , Animal Feed/analysis , Diet/veterinary , Hepatopancreas/immunology , Hepatopancreas/metabolism , Probiotics/administration & dosage , Probiotics/pharmacology , Dietary Supplements/analysis , Fermentation , Random Allocation , Gastrointestinal Microbiome/drug effects , Immunity, Innate , Dose-Response Relationship, Drug
17.
World J Microbiol Biotechnol ; 40(7): 204, 2024 May 17.
Article En | MEDLINE | ID: mdl-38755413

Globally colorectal cancer ranks as the third most widespread disease and the third leading cause of cancer-associated mortality. Immunotherapy treatments like PD-L1 blockade have been used to inhibit the PD-L1 legend, which boosts the activity of cytotoxic T lymphocytes. Recently, studies suggest that some probiotics could potentially enhance the effectiveness of immunotherapy treatments for cancer patients. We found that in Caco-2 and HT-29 cells, the live Leuconostoc mesenteroides treatment resulted an increase in the PD-L1 expression and this treatment stimulated interferon-gamma (IFN-γ) production in Jurkat T-cells. Due to the well-established ability of IFN-γ to enhance PD-L1 expression, the combination of IFN-γ and L. mesenteroides was used in colon cancer cell lines and a resulting remarkable increase of over tenfold in PD-L1 expression was obtained. Interestingly, when L. mesenteroides and IFN-γ are present, the blockage of PD-L1 using PD-L1 antibodies not only improved the viability of Jurkat T-cells but also significantly boosted the levels of IFN-γ and IL-2, the T-cells activation marker cytokines. In addition to upregulating PD-L1, L. mesenteroides also activated Toll-like receptors (TLRs) and NOD-like receptors (NODs) pathways, specifically through TLR2 and NOD2, while also exerting a suppressive effect on autophagy in colon cancer cell lines. In conclusion, our findings demonstrate a significant upregulation of PD-L1 expression in colon cancer cells upon co-culturing with L. mesenteroides. Moreover, the presence of PD-L1 antibodies during co-culturing activates Jurkat T cells. The observed enhancement in PD-L1 expression may be attributed to the inhibition of the Autophagy pathway or activation of the hippo pathway. KEY POINTS: Co-culturing L. mesenteroides increases PD-L1 gene and protein transaction in colon cancer. L. mesenteroides existing enhances T cells viability and activity. GPCR41/42 is a possible link between L. mesenteroides, YAP-1 and PD-L1.


B7-H1 Antigen , Colonic Neoplasms , Interferon-gamma , Leuconostoc mesenteroides , Up-Regulation , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Interferon-gamma/metabolism , Colonic Neoplasms/immunology , HT29 Cells , Jurkat Cells , Caco-2 Cells , Leuconostoc mesenteroides/metabolism , Leuconostoc mesenteroides/genetics , Interleukin-2/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Probiotics/pharmacology , Cell Line, Tumor , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics
18.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article En | MEDLINE | ID: mdl-38732016

Enterococcus spp. are normal intestinal tract microflorae found in poultry. However, the last decades have shown that several species, e.g., Enterococcus cecorum, have become emerging pathogens in broilers and may cause numerous losses in flocks. In this study, two combinations (H1 and H2) of menthol, 1,8-cineol, linalool, methyl salicylate, γ-terpinene, p-cymene, trans-anethole, terpinen-4-ol and thymol were used in an in vitro model, analyzing its effectiveness against the strains E. cecorum, E. faecalis, E. faecium, E. hirae and E. gallinarum isolated from broiler chickens from industrial farms. To identify the isolated strains classical microbiological methods and VITEK 2 GP cards were used. Moreover for E. cecorum a PCR test was used.. Antibiotic sensitivity (MIC) tests were performed for all the strains. For the composition H1, the effective dilution for E. cecorum and E. hirae strains was 1:512, and for E. faecalis, E. faecium and E. gallinarum, 1:1024. The second mixture (H2) showed very similar results with an effectiveness at 1:512 for E. cecorum and E. hirae and 1:1024 for E. faecalis, E. faecium and E. gallinarum. The presented results suggest that the proposed composition is effective against selected strains of Enterococcus in an in vitro model, and its effect is comparable to classical antibiotics used to treat this pathogen in poultry. This may suggest that this product may also be effective in vivo and provide effective support in the management of enterococcosis in broiler chickens.


Anti-Bacterial Agents , Chickens , Enterococcus , Microbial Sensitivity Tests , Animals , Chickens/microbiology , Enterococcus/drug effects , Enterococcus/genetics , Enterococcus/isolation & purification , Anti-Bacterial Agents/pharmacology , Poultry Diseases/microbiology , Poultry Diseases/drug therapy , Probiotics/pharmacology , Gram-Positive Bacterial Infections/veterinary , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/drug therapy
19.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38732060

The human gut microbiota, an intricate ecosystem within the gastrointestinal tract, plays a pivotal role in health and disease. Prebiotics, non-digestible food ingredients that beneficially affect the host by selectively stimulating the growth and/or activity of beneficial microorganisms, have emerged as a key modulator of this complex microbial community. This review article explores the evolution of the prebiotic concept, delineates various types of prebiotics, including fructans, galactooligosaccharides, xylooligosaccharides, chitooligosaccharides, lactulose, resistant starch, and polyphenols, and elucidates their impact on the gut microbiota composition. We delve into the mechanisms through which prebiotics exert their effects, particularly focusing on producing short-chain fatty acids and modulating the gut microbiota towards a health-promoting composition. The implications of prebiotics on human health are extensively reviewed, focusing on conditions such as obesity, inflammatory bowel disease, immune function, and mental health. The review further discusses the emerging concept of synbiotics-combinations of prebiotics and probiotics that synergistically enhance gut health-and highlights the market potential of prebiotics in response to a growing demand for functional foods. By consolidating current knowledge and identifying areas for future research, this review aims to enhance understanding of prebiotics' role in health and disease, underscoring their importance in maintaining a healthy gut microbiome and overall well-being.


Gastrointestinal Microbiome , Prebiotics , Humans , Probiotics/pharmacology , Obesity/microbiology , Obesity/diet therapy , Obesity/metabolism , Fatty Acids, Volatile/metabolism , Animals , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/diet therapy
20.
FASEB J ; 38(10): e23669, 2024 May 31.
Article En | MEDLINE | ID: mdl-38747734

Amomum xanthioides (AX) has been used as an edible herbal medicine to treat digestive system disorders in Asia. Additionally, Lactobacillus casei is a well-known probiotic commonly used in fermentation processes as a starter. The current study aimed to investigate the potential of Lactobacillus casei-fermented Amomum xanthioides (LAX) in alleviating metabolic disorders induced by high-fat diet (HFD) in a mouse model. LAX significantly reduced the body and fat weight, outperforming AX, yet without suppressing appetite. LAX also markedly ameliorated excessive lipid accumulation and reduced inflammatory cytokine (IL-6) levels in serum superior to AX in association with UCP1 activation and adiponectin elevation. Furthermore, LAX noticeably improved the levels of fasting blood glucose, serum insulin, and HOMA-IR through positive regulation of glucose transporters (GLUT2, GLUT4), and insulin receptor gene expression. In conclusion, the fermentation of AX demonstrates a pronounced mitigation of overnutrition-induced metabolic dysfunction, including hyperlipidemia, hyperglycemia, hyperinsulinemia, and obesity, compared to non-fermented AX. Consequently, we proposed that the fermentation of AX holds promise as a potential candidate for effectively ameliorating metabolic disorders.


Amomum , Diet, High-Fat , Fermentation , Lacticaseibacillus casei , Obesity , Animals , Diet, High-Fat/adverse effects , Mice , Obesity/metabolism , Male , Lacticaseibacillus casei/metabolism , Amomum/chemistry , Mice, Inbred C57BL , Probiotics/pharmacology , Uncoupling Protein 1/metabolism , Insulin Resistance , Mice, Obese , Adiponectin/metabolism , Insulin/metabolism , Insulin/blood , Blood Glucose/metabolism
...