Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.290
Filter
1.
Braz J Biol ; 84: e282609, 2024.
Article in English | MEDLINE | ID: mdl-39319927

ABSTRACT

Candida albicans is often associated with oral candidiasis, and drug-resistance profiles have contributed to an increase in morbidity and mortality. It is known that Lactobacillus spp. acts by competing for adhesion to the epithelium, absorption of nutrients and modulation of the human microbiota. Therefore, they are important to assist in the host's microbiological balance and reduce the growth of Candida spp. Until now, there have been no reports in the literature of reviews correlating to the use of Lactobacillus spp. in the treatment of oral candidiasis. Thus, this review aims to highlight the mechanisms of action of Lactobacillus spp. and methods that can be used in the treatment of oral candidiasis. This is a study carried out through the databases PubMed Central and Scientific Electronic Library Online, using the following keywords: Oral Candidiasis and Lactobacillus. Original articles about oral candidiasis were included, with both in vitro and in vivo analyses, and published from 2012 to 2022. Lactobacillus rhamnosus was the most common microorganism used in the experiments against Candida, acting mainly in the reduction of biofilm, filamentation, and competing for adhesion sites of Candida spp. Among in vivo studies, most researchers used immunosuppressed mouse modelsof Candida infection. The studies showed that Lactobacillus has a great potential as a probiotic, acting mainly in the prevention and treatment of mucosal diseases. Thus, the use of Lactobacillus may be a good strategy for the treatment of oral candidiasis.


Subject(s)
Candidiasis, Oral , Lactobacillus , Probiotics , Candidiasis, Oral/microbiology , Candidiasis, Oral/therapy , Candidiasis, Oral/drug therapy , Humans , Probiotics/therapeutic use , Lactobacillus/physiology , Animals , Biofilms/drug effects , Mice , Candida albicans/physiology
2.
Bull Exp Biol Med ; 177(4): 476-481, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39264564

ABSTRACT

The species identity of the studied lactobacillus strains was confirmed by matrix-activated laser desorption/ionization with time-of-flight ion separation (MALDI-TOF mass spectrometry). Lactobacillus strains differed in the dynamics of lactic acid accumulation and changes in the pH of the culture medium. The culture medium affected adhesion ability of lactobacilli. The ability to adhere does not affect the formation of biofilms by lactobacillus strains except for the L. acidophilus La5 strain, which has low adhesion ability and fewer microbial cells detected after mechanical destruction of the biofilm. The metabiotics of the lactobacillus culture medium have an antagonistic effect on conditionally pathogenic microorganisms. Adhesion, biofilm formation, and antagonistic activity of probiotic lactobacillus strains are strain-specific properties.


Subject(s)
Bacterial Adhesion , Biofilms , Lactobacillus , Probiotics , Biofilms/growth & development , Biofilms/drug effects , Probiotics/pharmacology , Bacterial Adhesion/drug effects , Lactobacillus/physiology , Hydrogen-Ion Concentration , Culture Media/chemistry , Lactic Acid/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Antibiosis/physiology , Lactobacillus acidophilus/physiology
3.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273118

ABSTRACT

The healthy cervicovaginal microbiota is dominated by various Lactobacillus species, which support a condition of eubiosis. Among their many functions, vaginal lactobacilli contribute to the maintenance of an acidic pH, produce antimicrobial compounds, and modulate the host immune response to protect against vaginal bacterial and fungal infections. Increasing evidence suggests that these beneficial bacteria may also confer protection against sexually transmitted infections (STIs) caused by viruses such as human papillomavirus (HPV), human immunodeficiency virus (HIV) and herpes simplex virus (HSV). Viral STIs pose a substantial public health burden globally, causing a range of infectious diseases with potentially severe consequences. Understanding the molecular mechanisms by which lactobacilli exert their protective effects against viral STIs is paramount for the development of novel preventive and therapeutic strategies. This review aims to provide more recent insights into the intricate interactions between lactobacilli and viral STIs, exploring their impact on the vaginal microenvironment, host immune response, viral infectivity and pathogenesis, and highlighting their potential implications for public health interventions and clinical management strategies.


Subject(s)
Lactobacillus , Vagina , Humans , Female , Lactobacillus/physiology , Vagina/microbiology , Vagina/virology , Vagina/immunology , Sexually Transmitted Diseases, Viral/immunology , Sexually Transmitted Diseases, Viral/prevention & control , Sexually Transmitted Diseases, Viral/virology , Microbiota
4.
Cell ; 187(19): 5393-5412.e30, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39121857

ABSTRACT

Negative psychological states impact immunity by altering the gut microbiome. However, the relationship between brain states and microbiome composition remains unclear. We show that Brunner's glands in the duodenum couple stress-sensitive brain circuits to bacterial homeostasis. Brunner's glands mediated the enrichment of gut Lactobacillus species in response to vagus nerve stimulation. Cell-specific ablation of the glands markedly suppressed Lactobacilli counts and heightened vulnerability to infection. In the forebrain, we mapped a vagally mediated, polysynaptic circuit connecting the central nucleus of the amygdala to Brunner's glands. Chronic stress suppressed central amygdala activity and phenocopied the effects of gland lesions. Conversely, excitation of either the central amygdala or parasympathetic vagal neurons activated Brunner's glands and reversed the effects of stress on the gut microbiome and immunity. The findings revealed a tractable brain-body mechanism linking psychological states to host defense.


Subject(s)
Duodenum , Gastrointestinal Microbiome , Stress, Psychological , Vagus Nerve , Animals , Mice , Duodenum/microbiology , Vagus Nerve/physiology , Male , Mice, Inbred C57BL , Amygdala/physiology , Lactobacillus/physiology , Neurons/metabolism
5.
Sci Rep ; 14(1): 15387, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965339

ABSTRACT

Probiotics offer a promising prophylactic approach against various pathogens and represent an alternative strategy to combat biofilm-related infections. In this study, we isolated vaginal commensal microbiota from 54 healthy Indian women to investigate their probiotic traits. We primarily explored the ability of cell-free supernatant (CFS) from Lactobacilli to prevent Uropathogenic Escherichia coli (UPEC) colonization and biofilm formation. Our findings revealed that CFS effectively reduced UPEC's swimming and swarming motility, decreased cell surface hydrophobicity, and hindered matrix production by downregulating specific genes (fimA, fimH, papG, and csgA). Subsequent GC-MS analysis identified Tryptamine, a monoamine compound, as the potent bioactive substance from Lactobacilli CFS, inhibiting UPEC biofilms with an MBIC of 4 µg/ml and an MBEC of 8 µg/ml. Tryptamine induced significant changes in E. coli colony biofilm morphology, transitioning from the Red, Dry, and Rough (RDAR) to the Smooth and White phenotype, indicating reduced extracellular matrix production. Biofilm time-kill assays demonstrated a four-log reduction in UPEC viability when treated with Tryptamine, highlighting its potent antibacterial properties, comparable to CFS treatment. Biofilm ROS assays indicated a significant elevation in ROS generation within UPEC biofilms, suggesting a potential antibacterial mechanism. Gene expression studies with Tryptamine-treated samples showed a reduction in expression of curli gene (csgA), consistent with CFS treatment. This study underscores the potential of Tryptamine from probiotic Lactobacilli CFS as a promising antibiofilm agent against UPEC biofilms.


Subject(s)
Biofilms , Lactobacillus , Probiotics , Tryptamines , Uropathogenic Escherichia coli , Vagina , Biofilms/drug effects , Biofilms/growth & development , Humans , Tryptamines/pharmacology , Female , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/physiology , Probiotics/pharmacology , Vagina/microbiology , Lactobacillus/drug effects , Lactobacillus/metabolism , Lactobacillus/physiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/prevention & control , Adult , Anti-Bacterial Agents/pharmacology
6.
Curr Microbiol ; 81(7): 202, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829392

ABSTRACT

There are massive sources of lactic acid bacteria (LAB) in traditional dairy products. Some of these indigenous strains could be novel probiotics with applications in human health and supply the growing needs of the probiotic industry. In this work, were analyzed the probiotic and technological properties of three Lactobacilli strains isolated from traditional Brazilian cheeses. In vitro tests showed that the three strains are safe and have probiotic features. They presented antimicrobial activity against pathogenic bacteria, auto-aggregation values around 60%, high biofilm formation properties, and a survivor of more than 65% to simulated acid conditions and more than 100% to bile salts. The three strains were used as adjunct cultures separately in a pilot-scale production of Prato cheese. After 45 days of ripening, the lactobacilli counts in the cheeses were close to 8 Log CFU/g, and was observed a reduction in the lactococci counts (around -3 Log CFU/g) in a strain-dependent manner. Cheese primary and secondary proteolysis were unaffected by the probiotic candidates during the ripening, and the strains showed no lipolytic effect, as no changes in the fatty acid profile of cheeses were observed. Thus, our findings suggest that the three strains evaluated have probiotic properties and have potential as adjunct non-starter lactic acid bacteria (NSLAB) to improve the quality and functionality of short-aged cheeses.


Subject(s)
Cheese , Probiotics , Cheese/microbiology , Brazil , Food Microbiology , Lactobacillus/metabolism , Lactobacillus/physiology , Lactobacillales/physiology , Lactobacillales/isolation & purification , Lactobacillales/metabolism , Lactobacillales/classification , Biofilms/growth & development , Fatty Acids/metabolism , Fermentation , Bile Acids and Salts/metabolism
7.
J Appl Microbiol ; 135(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38857885

ABSTRACT

AIMS: Klebsiella pneumoniae, an important opportunistic pathogen of nosocomial inflection, is known for its ability to form biofilm. The purpose of the current study is to assess how co- or mono-cultured probiotics affect K. pneumoniae's ability to produce biofilms and investigate the potential mechanisms by using a polyester nonwoven chemostat and a Caco-2 cell line. METHODS AND RESULTS: Compared with pure cultures of Lactobacillus rhamnosus and Lactobacillus sake, the formation of K. pneumoniae biofilm was remarkably inhibited by the mixture of L. rhamnosus, L. sake, and Bacillus subtilis at a ratio of 5:5:1 by means of qPCR and FISH assays. In addition, Lactobacillus in combination with B. subtilis could considerably reduce the adherence of K. pneumoniae to Caco-2 cells by using inhibition, competition, and displacement assays. According to the RT-PCR assay, the adsorption of K. pneumoniae to Caco-2 cells was effectively inhibited by the co-cultured probiotics, leading to significant reduction in the expression of proinflammatory cytokines induced by K. pneumoniae. Furthermore, the HPLC and RT-PCR analyses showed that the co-cultured probiotics were able to successfully prevent the expression of the biofilm-related genes of K. pneumoniae by secreting plenty of organic acids as well as the second signal molecule (c-di-GMP), resulting in inhibition on biofilm formation. CONCLUSION: Co-culture of L. sake, L. rhamnosus, and B. subtilis at a ratio of 5:5:1 could exert an antagonistic effect on the colonization of pathogenic K. pneumoniae by down-regulating the expression of biofilm-related genes. At the same time, the co-cultured probiotics could effectively inhibit the adhesion of K. pneumoniae to Caco-2 cells and block the expression of proinflammatory cytokines induced by K. pneumoniae.


Subject(s)
Biofilms , Coculture Techniques , Klebsiella pneumoniae , Probiotics , Biofilms/growth & development , Klebsiella pneumoniae/physiology , Humans , Probiotics/pharmacology , Caco-2 Cells , Bacillus subtilis/physiology , Bacillus subtilis/genetics , Lacticaseibacillus rhamnosus/physiology , Bacterial Adhesion , Lactobacillus/physiology , Cytokines/metabolism
8.
Sci Rep ; 14(1): 9365, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38654026

ABSTRACT

Strategies against the opportunistic fungal pathogen Candida albicans based on probiotic microorganisms represent a promising alternative to traditional antifungals. Here, we investigated the effects of Lactobacillaceae isolates from fermented foods or the human vagina, alone or in combination with the probiotic yeast Saccharomyces cerevisiae CNCM I-3856, against C. albicans in vitro. Nine out of nineteen tested strains of Lactobacillaceae inhibited growth of C. albicans with inhibition zones of 1-3 mm in spot assays. Five out of nineteen lactobacilli tested as such or in combination with S. cerevisiae CNCM I-3856 also significantly inhibited C. albicans hyphae formation, including Limosilactobacillus fermentum LS4 and L. fermentum LS5 resulting in respectively 62% and 78% hyphae inhibition compared to the control. Thirteen of the tested nineteen lactobacilli aggregated with the yeast form of C. albicans, with Lactiplantibacillus carotarum AMBF275 showing the strongest aggregation. The aggregation was enhanced when lactobacilli were combined with S. cerevisiae CNCM I-3856. No significant antagonistic effects were observed between the tested lactobacilli and S. cerevisiae CNCM I-3856. The multifactorial activity of Lactobacillaceae strains alone or combined with the probiotic S. cerevisiae CNCM I-3856 against C. albicans without antagonistic effects between the beneficial strains, paves the way for developing consortium probiotics for in vivo applications.


Subject(s)
Candida albicans , Lactobacillus , Probiotics , Saccharomyces cerevisiae , Candida albicans/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/drug effects , Probiotics/pharmacology , Lactobacillus/physiology , Humans , Hyphae/drug effects , Hyphae/growth & development , Antibiosis , Female , Vagina/microbiology
9.
PLoS One ; 19(4): e0301822, 2024.
Article in English | MEDLINE | ID: mdl-38603764

ABSTRACT

The human gut is a complex environment where the microbiota and its metabolites play a crucial role in the maintenance of a healthy state. The aim of the present work is the reconstruction of a new in vitro minimal human gut microbiota resembling the microbe-microbe networking comprising the principal phyla (Bacillota, Bacteroidota, Pseudomonadota, and Actinomycetota), to comprehend the intestinal ecosystem complexity. In the reductionist model, we mimicked the administration of Maitake extract as prebiotic and a probiotic formulation (three strains belonging to Lactobacillus and Bifidobacterium genera), evaluating the modulation of strain levels, the release of beneficial metabolites, and their health-promoting effects on human cell lines of the intestinal environment. The administration of Maitake and the selected probiotic strains generated a positive modulation of the in vitro bacterial community by qPCR analyses, evidencing the prominence of beneficial strains (Lactiplantibacillus plantarum and Bifidobacterium animalis subsp. lactis) after 48 hours. The bacterial community growths were associated with the production of metabolites over time through GC-MSD analyses such as lactate, butyrate, and propionate. Their effects on the host were evaluated on cell lines of the intestinal epithelium and the immune system, evidencing positive antioxidant (upregulation of SOD1 and NQO1 genes in HT-29 cell line) and anti-inflammatory effects (production of IL-10 from all the PBMCs). Therefore, the results highlighted a positive modulation induced by the synergic activities of probiotics and Maitake, inducing a tolerogenic microenvironment.


Subject(s)
Bifidobacterium animalis , Gastrointestinal Microbiome , Grifola , Probiotics , Humans , Ecosystem , Intestinal Mucosa/microbiology , Lactobacillus/physiology , Probiotics/pharmacology
10.
Vet Med Sci ; 10(3): e1463, 2024 05.
Article in English | MEDLINE | ID: mdl-38659354

ABSTRACT

BACKGROUND: The most prevalent probiotic bacterium employed in the food industry is Lactobacillus because it can produce metabolites with antibacterial capabilities and exhibits hostility towards infections and microorganisms that cause spoilage. AIM: This study set out to identify naturally occurring Lactobacillus and plantaricin (pln EF) coding genes in raw cow milk and to assess the antibacterial potency of isolated Lactobacillus isolates. METHODS: Following enrichment in De Man, Rogosa and Sharpe (MRS) broth, single colonies were isolated, and pure colonies were obtained by streaking on MRS agar. The 16S rRNA gene was amplified using polymerase chain reaction (PCR) to confirm the cultural positivity of all isolates. Additionally, the presence of plantaricin was verified by targeting the pln EF gene through PCR. OUTCOME: Out of the 166 raw milk specimens acquired from cows, 153 (91.17%; CI: 86.98-95.76) were identified as positive for Lactobacillus through both culture and biochemical screening. Subsequently, 121 (72.89%; CI: 65.46-79.49) of the isolates were affirmed to harbour Lactobacillus through PCR analysis. Within this subset, 6 isolates (4.96%; CI: 1.84-10.48) were found to possess the plnEF gene. When exposed to Lactobacillus isolates, Salmonella Typhimurium and Salmonella enterica displayed an average maximum zone of inhibition with a diameter measuring 24 mm. In contrast, Escherichia coli exhibited an average minimum zone of inhibition, featuring a diameter of 11 mm. Additionally, the Lactobacillus isolates demonstrated inhibitory zones against Staphylococcus aureus, Klebsiella pneumoniae and Klebsiella oxytoca, measuring 14, 22 and 19 mm, respectively. CLINICAL SIGNIFICANCE: Lactic acid bacteria, particularly Lactobacilli, are plentiful in cow milk and possess broad-spectrum antibacterial properties.


Subject(s)
Lactobacillus , Milk , Milk/microbiology , Animals , Cattle , Lactobacillus/genetics , Lactobacillus/physiology , Lactobacillus/isolation & purification , Bangladesh/epidemiology , Anti-Bacterial Agents/pharmacology , Female , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics
11.
Benef Microbes ; 15(3): 293-310, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38677716

ABSTRACT

Oral administration of probiotics has been proposed as a promising biotherapy to prevent and treat different diseases related to gastrointestinal disorders, such as irritable bowel syndrome (IBS). Due to the increasing research area on the characterisation of new probiotic bacterial strains, it is necessary to perform suitable in vitro experiments, using pertinent cellular models, in order to establish appropriate readout profiles based on IBS symptoms and subtypes. In this work, a collection of 30 candidate strains, belonging mainly to the Lactobacillus and Bifidobacterium genera, were screened using three different sets of in vitro experiments with different readouts to identify promising probiotic strains with: (1) the ability to inhibit the synthesis of IL-8 production by TNF-α stimulated HT-29 cells, (2) immunomodulatory properties quantified as increased IL-10 levels in peripheral blood mononuclear cell (PBMCs), and (3) the ability to maintain epithelial barrier integrity by increasing the trans-epithelial/endothelial electrical resistance (TEER) values in Caco-2 cells. Based on these criteria, three strains were selected: Lactobacillus gasseri PI41, Lacticaseibacillus rhamnosus PI48 and Bifidobacterium animalis subsp. lactis PI50, and tested in a murine model of low-grade inflammation induced by dinitrobenzene sulfonic acid (DNBS), which mimics some of the symptoms of IBS. Among the three strains, L. gasseri PI41 improved overall host well-being by preventing body weight loss in DNBS-treated mice and restored gut homeostasis by normalising the intestinal permeability and reducing pro-inflammatory markers. Therefore, the potential of this strain was confirmed in a second murine model known to reproduce IBS symptoms: the neonatal maternal separation (NMS) model. The PI41 strain was effective in preventing intestinal permeability and reducing colonic hypersensitivity. In conclusion, the set of in vitro experiments combined with in vivo assessments allowed us to identify a promising probiotic candidate strain, L. gasseri PI41, in the context of IBS.


Subject(s)
Irritable Bowel Syndrome , Probiotics , Probiotics/administration & dosage , Probiotics/pharmacology , Irritable Bowel Syndrome/therapy , Irritable Bowel Syndrome/microbiology , Humans , Animals , Mice , Caco-2 Cells , HT29 Cells , Disease Models, Animal , Leukocytes, Mononuclear/immunology , Lactobacillus/physiology , Interleukin-8/metabolism , Bifidobacterium/physiology , Interleukin-10 , Lactobacillus gasseri , Lacticaseibacillus rhamnosus/physiology , Male , Bifidobacterium animalis/physiology
12.
Poult Sci ; 103(6): 103650, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555756

ABSTRACT

Dietary ingredient and nutrient composition may affect the efficacy of additives in broilers. Specific feed ingredients can represent dietary challenging conditions for broilers, resulting in impaired performances and health, which might be alleviated by dietary probiotics and postbiotics. We assessed the effects of a Lactobacilli probiotic (Pro) and postbiotic (Post) when added to a standard (SD) and challenge (CD) diet. A completely randomized block study with 2 diets (SD, CD) and 3 additive conditions (Control, Pro and Post) involving 1,368 one-day-old Ross male broilers, equally distributed among 36 pens, from d1 to d42 was conducted. Both diets were formulated to contain identical levels of nutrients, with CD formulated to be richer than SD in nonstarch polysaccharides using rye and barley as ingredients. Readout parameters included growth performance parameters, footpad lesions score, blood minerals and biochemical parameters, and tibia health, strength, and composition. Compared to SD, CD decreased BW (1,936 vs. 2,033 g; p = 0.001), increased FCR (p < 0.01) and impaired tibia health and strength (p < 0.05) at d35, thereby confirming the challenging effect of CD. Pro and Post increased BW in CD (+4.7 and +3.2%, respectively, at d35; P < 0.05) but not in the SD group, without affecting FCR. Independently of the diet, Pro increased plasma calcium, phosphorus and uric acid at d21 (+6.2, +7.4, and +15.5%, respectively) and d35 (+6.6, +6.2 and +21.0%, respectively) (P < 0.05) while Post increased plasma magnesium only at d21 (+11.3%; P = 0.037). Blood bile acids were affected by additives in an age- and diet-dependent manner, with some opposite effects between dietary conditions. Diet composition modulated Pro and Post effects on broiler growth performance. Additionally, Pro and Post affected animal metabolism and leg health diet-dependently for some but not all investigated parameters. Our findings show that the effects of pro- and postbiotics on the growth performance and physiology of broilers can be dependent on diet composition and thus possibly other factors affecting diet characteristics.


Subject(s)
Animal Feed , Animal Nutritional Physiological Phenomena , Chickens , Diet , Probiotics , Random Allocation , Animals , Chickens/growth & development , Chickens/physiology , Probiotics/administration & dosage , Probiotics/pharmacology , Male , Diet/veterinary , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Dietary Supplements/analysis , Lactobacillus/physiology
13.
Methods Mol Biol ; 2763: 353-358, 2024.
Article in English | MEDLINE | ID: mdl-38347425

ABSTRACT

The ability of Lactobacillus to adhere to mucin is a parameter for evaluating the effectiveness of probiotics. In particular, a competitive inhibition assay of pathogenic bacteria using mucin-adherent lactobacilli is useful for identifying Lactobacillus strains capable of preventing mucus from being colonized by pathogens. Here, we describe an adhesion inhibition assay method for Helicobacter pylori to porcine gastric mucin by Limosilactobacillus reuteri.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Probiotics , Animals , Swine , Lactobacillus/physiology , Mucins , Bacterial Adhesion/physiology
14.
Nutrients ; 16(4)2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38398870

ABSTRACT

Several billion microorganisms reside in the gastrointestinal lumen, including viruses, bacteria, fungi, and yeast. Among them, probiotics were primarily used to cure digestive disorders such as intestinal infections and diarrhea; however, with a paradigm shift towards alleviating health through food, their importance is large. Moreover, recent studies have changed the perspective that probiotics prevent numerous ailments in the major organs. Probiotics primarily produce biologically active compounds targeting discommodious pathogens. This review demonstrates the implications of using probiotics from different genres to prevent and alleviate ailments in the primary human organs. The findings reveal that probiotics immediately activate anti-inflammatory mechanisms by producing anti-inflammatory cytokines such as interleukin (IL)-4, IL-10, IL-11, and IL-13, and hindering pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α by involving regulatory T cells (Tregs) and T helper cells (Th cells). Several strains of Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus casei, Lactobacillus reuteri, Bifidobacterium longum, and Bifidobacterium breve have been listed among the probiotics that are excellent in alleviating various simple to complex ailments. Therefore, the importance of probiotics necessitates robust research to unveil the implications of probiotics, including the potency of strains, the optimal dosages, the combination of probiotics, their habitat in the host, the host response, and other pertinent factors.


Subject(s)
Lacticaseibacillus casei , Probiotics , Humans , Lactobacillus/physiology , Cytokines , Probiotics/therapeutic use , Anti-Inflammatory Agents/pharmacology
15.
Folia Microbiol (Praha) ; 69(4): 927-939, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38308067

ABSTRACT

Helicobacter pylori infection is the major risk factor associated with the development of gastric cancer. Currently, administration of standard antibiotic therapy combined with probiotics and postbiotics has gained significant attention in the management of H. pylori infection. In this work, the immunomodulatory effects of Lactobacillus crispatus-derived extracellular vesicles (EVs) and cell-free supernatant (CFS) were investigated on H. pylori-induced inflammatory response in human gastric adenocarcinoma (AGS) cells. L. crispatus-derived EVs were isolated by ultracentrifugation and physically characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM). Furthermore, the protein content of L. crispatus-derived EVs was also evaluated by SDS-PAGE. Cell viability of AGS cells exposed to varying concentrations of EVs and CFS was assessed by MTT assay. The mRNA expression of IL-1ß, IL-6, IL-8, TNF-α, IL-10, and TGF-ß genes was determined by RT-qPCR. ELISA was used for the measurement of IL-8 production in AGS cells. In addition, EVs (50 µg/mL) and CFS modulated the H. pylori-induced inflammation by downregulating the mRNA expression of IL-1ß, IL-6, IL-8, and TNF-α, and upregulating the expression of IL-10, and TGF-ß genes in AGS cells. Furthermore, H. pylori-induced IL-8 production was dramatically decreased after treatment with L. crispatus-derived EVs and CFS. In conclusion, our observation suggests for the first time that EVs released by L. crispatus strain RIGLD-1 and its CFS could be recommended as potential therapeutic agents against H. pylori-triggered inflammation.


Subject(s)
Cytokines , Epithelial Cells , Extracellular Vesicles , Helicobacter pylori , Humans , Helicobacter pylori/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry , Extracellular Vesicles/immunology , Epithelial Cells/microbiology , Cytokines/metabolism , Cytokines/genetics , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Helicobacter Infections/microbiology , Helicobacter Infections/immunology , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Lactobacillus/metabolism , Lactobacillus/physiology , Inflammation/microbiology , Probiotics/pharmacology
16.
J Microbiol Biotechnol ; 34(4): 854-862, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38326923

ABSTRACT

Lactobacillus is a commonly used probiotic, and many researchers have focused on its stress response to improve its functionality and survival. However, studies on persister cells, dormant cells that aid bacteria in surviving general stress, have focused on pathogenic bacteria that cause infection, not Lactobacillus. Thus, understanding Lactobacillus persister cells will provide essential clues for understanding how Lactobacillus survives and maintains its function under various environmental conditions. We treated Lactobacillus strains with various antibiotics to determine the conditions required for persister formation using kill curves and transmission electron microscopy. In addition, we observed the resuscitation patterns of persister cells using single-cell analysis. Our results show that Lactobacillus creates a small population of persister cells (0.0001-1% of the bacterial population) in response to beta-lactam antibiotics such as ampicillin and amoxicillin. Moreover, only around 0.5-1% of persister cells are heterogeneously resuscitated by adding fresh media; the characteristics are typical of persister cells. This study provides a method for forming and verifying the persistence of Lactobacillus and demonstrates that antibiotic-induced Lactobacillus persister cells show characteristics of dormancy, sensitivity of antibiotics, same as exponential cells, multi-drug tolerance, and resuscitation, which are characteristics of general persister cells. This study suggests that the mechanisms of formation and resuscitation may vary depending on the characteristics, such as the membrane structure of the bacterial species.


Subject(s)
Ampicillin , Anti-Bacterial Agents , Lactobacillus , Microbial Sensitivity Tests , Microbial Viability , Anti-Bacterial Agents/pharmacology , Lactobacillus/physiology , Ampicillin/pharmacology , Microbial Viability/drug effects , Microscopy, Electron, Transmission , Probiotics , Amoxicillin/pharmacology
17.
Br J Pharmacol ; 181(1): 162-179, 2024 01.
Article in English | MEDLINE | ID: mdl-37594378

ABSTRACT

BACKGROUND AND PURPOSE: Membranous nephropathy (MN) is an immune-mediated glomerular disease in adults. Antibody- and antigen-bonding mechanisms have been largely clarified, but the subepithelium immune complex deposition-mediated downstream molecular mechanisms are currently unresolved. Increasing evidence has suggested that gut microbiota contribute to MN pathogenesis. EXPERIMENTAL APPROACH: In this study, we identified alterations in faecal gut microbiota and serum metabolites that mediate an aryl hydrocarbon receptor (AhR) mechanism in cationic bovine serum albumin (CBSA)-induced MN rats and in patients with idiopathic MN (IMN). KEY RESULTS: Impaired renal function correlated with the relative abundance of reduced faecal probiotics, Lactobacillus and Bifidobacterium, and altered serum levels of tryptophan-produced indole derivatives (TPIDs) in MN rats. Further results showed that reduced relative abundance of five probiotics, namely Lactobacillus johnsonii, Lactobacillus murinus, Lactobacillus vaginalis, Lactobacillus reuteri and Bifidobacterium animalis, positively correlated with decreased levels of indole-3-pyruvic acid, indole-3-aldehyde and tryptamine and negatively correlated with increased levels of indole-3-lactic acid and indole-3-acetic acid in serum of MN rats. Altered five probiotics and five TPIDs also were observed in patients with IMN. Further studies showed that MN rats exhibited a significant increase in intrarenal mRNA expression of AhR and its target genes CYP1A1, CYP1A2 and CYP1B1, which were accompanied by protein expression of down-regulated cytoplasmic AhR, but up-regulated nuclear AhR, in MN rats and IMN patients. CONCLUSION AND IMPLICATIONS: Activation of the intrarenal AhR signalling pathway may involve five TPIDs. These data suggest that gut microbiota could influence MN through TPIDs that engage host receptors.


Subject(s)
Gastrointestinal Microbiome , Glomerulonephritis, Membranous , Indoles , Lactobacillus , Receptors, Aryl Hydrocarbon , Lactobacillus/physiology , Glomerulonephritis, Membranous/microbiology , Tryptophan/pharmacology , Indoles/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Humans , Animals , Rats , Male , Rats, Sprague-Dawley , Signal Transduction
18.
Arch Razi Inst ; 78(3): 1115-1130, 2023 06.
Article in English | MEDLINE | ID: mdl-38028837

ABSTRACT

Typhoid fever is one of the most commonly disseminated diseases and is considered to be linked to poor sanitation. It is responsible for 2-5% of all deaths, and its causative agent is Salmonella typhi. The current study aimed to investigate the antibacterial activity of prebiotics (inulin and starch) and probiotics against multidrug resistance of S. typhi bacterial isolates. Determination of the inhibitory effect of probiotics and prebiotics against S. typhi isolates was performed by agar well diffusion method and minimal inhibitory concentration. Body samples of all eligible patients were collected and cultured. Finally, 50 (25%) out of the total cultured samples were S. Typhi bacteria isolated from different samples. The bacteria were mainly found in blood, followed by stool and fluid (74%, 24%, and 2%, respectively). On differential medium, xylose lysine deoxycholate agar, the colonies appear red with black centers, while on MacConkey agar, the colonies appear smooth, pale, transparent, colorless, and raised. Regarding the inhibition zone values of bacteriocins of Lactobacillus from Yogurt against S. typhi in plate, significant differences were identified between the ones with and without prebiotic addition. Accordingly, the value of the inhibition zone for those without prebiotic addition (13.18±7.403) was significantly lower than that of cutoff values of 20 with a significant difference of -6.820 (t= -6.514, df 49, P=0.000). Moreover, the inhibition effect of prebiotics (inulin and starch) against S. typhi at 37 °C for 24 h in part dish glucose as control, only the mean of inulin was found to be significantly lower than that of the cutoff value of 18 with the mean difference of -3.900 (t=-4.115, df 49, P=0.000). Other prebiotics of glucose and starch in 24 h showed negative inhibition. Probiotics are live microorganisms that have beneficial host effects by enhancing microbial balance in the intestine, whereas prebiotics are indigestible food components having beneficial effects by enhancing the activity and growth of one or more colonic bacteria. Lactobacillus filtrates had considerable effects against the test S. typhi isolates.


Subject(s)
Probiotics , Typhoid Fever , Humans , Prebiotics , Typhoid Fever/prevention & control , Inulin , Agar , Probiotics/pharmacology , Lactobacillus/physiology , Glucose
19.
Arch Razi Inst ; 78(3): 831-841, 2023 06.
Article in English | MEDLINE | ID: mdl-38028859

ABSTRACT

Probiotics have been used for over a century to prevent and treat diseases. They can reduce the effects of gastroenteritis and are now used to treat acute diarrhea. This study aimed to evaluate the co-aggregative effects of probiotics bacteria against diarrheal causative bacteria. For this purpose, 11 isolates of probiotic bacteria were used in the current study, including three Lactobacillus plantarum, one Lactobacillus gasseri, two Lactobacillus fermentum, three Lactobacillus acidophilus, and two Lactococcus garvieae isolates. All isolates were tested for antibiotic susceptibility, autoaggregation ability, adhesion ability, antibacterial activity, acid tolerance, and bile salts tolerance. The results showed that most of them had the ability to autoaggregate after 4 h, with the highest percentage of 57.14% for L. fermentum. For the antibiotic susceptibility test, all the isolates showed resistance against trimethoprim/sulfamethoxazole, except one isolate. Moreover, all the isolates, except one, were susceptible to both vancomycin and tetracycline. All tested isolates had adhesion ability with different survival rates, which reached 34.57% for L. plantarum in acidic conditions. Besides, the highest survival rate was 85.17%, which belonged to L. garvieae, for bile salt tolerance. Probiotic isolates had an antibacterial effect against diarrhea-causative bacteria with an inhibition diameter of 17-49 mm for different Lactobacillus spp. and Lactococcus spp. isolates. Furthermore, the co-aggregation ability of probiotic isolates against diarrhea-causative bacteria was studied, and results showed that probiotic isolates had a co-aggregative effect against diarrhea-causative bacteria, Escherichia coli, Shigella sonnei, and Providencia alcalifaciens, after 24 h of incubation. The highest co-aggregative effect of probiotics isolates belonged to L. fermentum and L. acidophilus against P. alcalifaciens with a co-aggregation percentage of 100%, while the lowest co-aggregation rate was 14.29% against E. coli. The findings revealed the probiotic properties and co-aggregative effects of probiotic bacteria against diarrhea-causative bacteria.


Subject(s)
Escherichia coli , Probiotics , Animals , Lactobacillus/physiology , Anti-Bacterial Agents/pharmacology , Bile Acids and Salts/pharmacology , Diarrhea , Probiotics/pharmacology
20.
Cells ; 12(21)2023 10 28.
Article in English | MEDLINE | ID: mdl-37947616

ABSTRACT

Probiotic bacteria belonging to Lactobacillus spp. are important producers of bioactive molecules, known as postbiotics, that play essential roles in the immunological support of the intestinal mucosa. In this study, the system of co-culture of intestinal epithelial cells with macrophage cells in vitro was used to study the potential effect of postbiotic fractions of L. rhamonosus and L. plantarum on the modulation of the immune response induced by pro-inflammatory stimuli. This study's results revealed that the presence of probiotic bacterial components on the mucosal surface in the early and late stage of inflammatory conditions is based on cellular interactions that control inflammation and consequent damage to the intestinal epithelium. In our studies, heat killed fractions of probiotic bacteria and their extracted proteins showed a beneficial effect on controlling inflammation, regardless of the strain tested, consequently protecting intestinal barrier damage. In conclusion, the presented results emphasize that the fractions of probiotic bacteria of L. plantarum and L. rhamnosus may play a significant role in the regulation of LPS-mediated cytotoxic activity in intestinal epithelial cells. The fractions of probiotic strains of L. rhamnosus and L. plantarum showed the potential to suppress inflammation, effectively activating the anti-inflammatory cytokine IL-10 and modulating the IL-18-related response.


Subject(s)
Lacticaseibacillus rhamnosus , Lactobacillus plantarum , Probiotics , Humans , Lactobacillus plantarum/physiology , Lactobacillus/physiology , Probiotics/pharmacology , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL