Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (177)

Search Parameters:
Keywords = next-generation probiotic

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 1463 KB  
Review
An Overview of Fish Disease Diagnosis and Treatment in Aquaculture in Bangladesh
by Md. Naim Mahmud, Abu Ayub Ansary, Farzana Yasmin Ritu, Neaz A. Hasan and Mohammad Mahfujul Haque
Aquac. J. 2025, 5(4), 18; https://doi.org/10.3390/aquacj5040018 (registering DOI) - 4 Oct 2025
Abstract
Aquaculture has rapidly become a vital sector for ensuring global food security by meeting the growing demand for animal protein. Bangladesh, one of the world’s leading aquaculture producers, recorded a production of 4.91 million MT in 2022–2023, largely driven by inland farming systems. [...] Read more.
Aquaculture has rapidly become a vital sector for ensuring global food security by meeting the growing demand for animal protein. Bangladesh, one of the world’s leading aquaculture producers, recorded a production of 4.91 million MT in 2022–2023, largely driven by inland farming systems. Despite this remarkable growth, the sector is highly vulnerable to disease outbreaks, which are aggravated by different factors. Pathogens such as bacteria, viruses, fungi, and parasites cause significant losses, while conventional disease diagnosis in Bangladesh still depends mainly on visual assessment and basic laboratory techniques, limiting early detection. This narrative review highlights recent advances in diagnostics as molecular tools, immunodiagnostics, nanodiagnostics, machine learning, and next-generation sequencing (NGS) that are widely applied globally but remain limited in Bangladesh due to infrastructure gaps, lack of skilled manpower, and resource constraints. Current treatment strategies largely rely on antibiotics and aquaculture medicinal products (AMPs), often misused without proper diagnosis, contributing to antimicrobial resistance (AMR). Promising alternatives, including probiotics, immunostimulants, vaccines, and enhanced biosecurity, require greater adoption and farmer awareness. The near-term priorities for Bangladesh include standardized disease and AMR surveillance, prudent antibiotic stewardship, phased adoption of validated rapid diagnostics, and investment in diagnostic and human capacity. Policy-level actions, including a national aquatic animal health strategy, stricter antimicrobial regulation, strengthening diagnostic infrastructure in institution, are crucial to achieve sustainable disease management and ensure long-term resilience of aquaculture in Bangladesh. Full article
Show Figures

Figure 1

21 pages, 1813 KB  
Article
A Comparison of the Response of the Human Intestinal Microbiota to Probiotic and Nutritional Interventions In Vitro and In Vivo—A Case Study
by Agnieszka Rudzka, Ondřej Patloka, Magdalena Płecha, Marek Zborowski, Tomasz Królikowski, Michał Oczkowski, Danuta Kołożyn-Krajewska, Marcin Kruk, Marcelina Karbowiak, Wioletta Mosiej and Dorota Zielińska
Nutrients 2025, 17(19), 3093; https://doi.org/10.3390/nu17193093 - 29 Sep 2025
Abstract
Background/Objectives: With increasing knowledge of the role of the microbiota in health and disease, the need for the reliable simulation of its behavior in response to various factors, such as diet and probiotic administration in in vitro conditions, has emerged. Although many studies [...] Read more.
Background/Objectives: With increasing knowledge of the role of the microbiota in health and disease, the need for the reliable simulation of its behavior in response to various factors, such as diet and probiotic administration in in vitro conditions, has emerged. Although many studies utilize developed systems, data on how accurately these systems represent individual microbiota responses are scarce. Methods: In the present study, the Simulator of Human Intestinal Microbial Ecosystem (SHIME®) was exposed to experimental conditions mimicking the application of probiotics and dietary changes in the study participant. Next-generation 16S rRNA sequencing was used to reveal the structure of the microbial communities in the analyzed samples. Results: Analysis of 17 samples revealed that predominantly diet and, to a lesser extent, probiotics had a divergent effect on the microbiota’s fluctuations dependent on the culture environment. Despite this, results from both in vitro and in vivo conditions aligned well with previously published data on the expected impact of dietary changes on the intestinal microbial community. Conclusions: The anecdotal evidence presented in this study suggested that current in vitro technology enables the reproduction of some of the microbiota responses that are well known from in vivo research. However, further work is required to enable simulations of an individual microbiota. Full article
(This article belongs to the Special Issue Effect of Dietary Components on Gut Homeostasis and Microbiota)
Show Figures

Figure 1

23 pages, 2800 KB  
Article
Genome-Driven Insights into Lactococcus sp. KTH0-1S Highlights Its Biotechnological Potential as a Cell Factory
by Nisit Watthanasakphuban, Hind Abibi, Nuttakan Nitayapat, Phitsanu Pinmanee, Chollachai Klaysubun, Nattarika Chaichana, Komwit Surachat and Suttipun Keawsompong
Biology 2025, 14(10), 1323; https://doi.org/10.3390/biology14101323 - 25 Sep 2025
Abstract
The safety, genetic distinctiveness, and functional capabilities of Lactococcus sp. KTH0-1S, a strain isolated from Thai fermented shrimp (Kung-Som), were investigated to assess its potential as a next-generation probiotic and microbial cell factory. Whole-genome sequencing and multilocus sequence typing (MLST) analysis revealed that [...] Read more.
The safety, genetic distinctiveness, and functional capabilities of Lactococcus sp. KTH0-1S, a strain isolated from Thai fermented shrimp (Kung-Som), were investigated to assess its potential as a next-generation probiotic and microbial cell factory. Whole-genome sequencing and multilocus sequence typing (MLST) analysis revealed that Lactococcus sp. KTH0-1S is a novel, phylogenetically distinct strain within the Lactococcus genus. Comprehensive in silico safety evaluation confirmed the absence of antimicrobial resistance genes and major virulence factors, supporting its suitability for food-grade applications. The genome encodes multiple probiotic-relevant traits, including stress tolerance (e.g., dnaK, clpP), adhesion and biofilm formation (e.g., gapA, luxS, glf2), and nutrient acquisition genes, enabling adaptation to gastrointestinal and fermentation environments. Notably, Lactococcus sp. KTH0-1S harbors a chromosomally encoded nisin Z biosynthesis gene cluster with auto-induction capability, providing a self-regulated and stable alternative to conventional plasmid-based NICE systems in Lactococcus lactis. The strain also exhibits nisin immunity, allowing tolerance to high nisin concentrations, thus supporting robust protein production. Genomic evidence and phenotypic assays confirmed a functional respiration metabolism activated by heme supplementation, enhancing biomass yield and culture stability. Furthermore, the presence of diverse CAZyme families (GHs, GTs, CEs) enables utilization of various carbohydrate substrates, including lignocellulosic and starchy agro-industrial residues. These properties collectively underscore Lactococcus sp. KTH0-1S as a safe, stable, and metabolically versatile candidate for probiotic applications and as a cost-effective, food-grade expression host for biotechnological production. Full article
Show Figures

Figure 1

30 pages, 1950 KB  
Review
Probiotics in Pet Food: A Decade of Research, Patents, and Market Trends
by Phatthranit Klinmalai, Pitiya Kamonpatana, Janenutch Sodsai, Atcharawan Srisa, Khwanchat Promhuad, Yeyen Laorenza, Attawit Kovitvadhi, Sathita Areerat, Anusorn Seubsai, Massalin Nakphaichit and Nathdanai Harnkarnsujarit
Foods 2025, 14(19), 3307; https://doi.org/10.3390/foods14193307 - 24 Sep 2025
Viewed by 85
Abstract
Increasing interest in functional nutrition has driven the incorporation of probiotics into pet food formulations to enhance digestive health, immune response, and overall well-being in companion animals. This systematic review examines scientific publications, patents, and market developments related to probiotic-enriched pet food from [...] Read more.
Increasing interest in functional nutrition has driven the incorporation of probiotics into pet food formulations to enhance digestive health, immune response, and overall well-being in companion animals. This systematic review examines scientific publications, patents, and market developments related to probiotic-enriched pet food from 2014 to 2024. We evaluate major probiotic taxa—including Lactobacillus spp., Bifidobacterium spp., Bacillus spp., and Saccharomyces cerevisiae—based on their resilience during processing, gastrointestinal survival, and documented health benefits. Delivery technologies such as microencapsulation, coating matrices, and post-processing supplementation are analyzed for their effectiveness in preserving probiotic viability within dry and wet food matrices. Patent landscape analysis highlights innovation trends in strain selection, formulation design, and processing methods. In vivo and in vitro studies demonstrate that probiotic supplementation modulates gut microbiota composition, improves fecal parameters, enhances immune markers, and promotes nutrient absorption in both canine and feline models. Market data reveal rapid expansion of commercial probiotic pet food products, yet scientific research remains limited compared to human nutrition. Overall, the findings indicate that while probiotics hold clear potential to improve gastrointestinal health and immunity in pets, evidence remains fragmented, particularly for cats and long-term outcomes. Bridging the gap between industrial innovation and controlled clinical validation will be essential for developing next-generation probiotic pet foods. Full article
(This article belongs to the Section Food Microbiology)
Show Figures

Figure 1

23 pages, 2338 KB  
Article
Candida intermedia Supplementation Enhances Immune Response and Modulates the Gut Microbiome in SARS-CoV-2 Vaccinated Mice
by Renan E. A. Piraine, Neida L. Conrad, Vitória S. Gonçalves, Jeferson V. Ramos, Júlia L. Froldi, Fausto Almeida and Fábio P. L. Leite
J. Fungi 2025, 11(9), 685; https://doi.org/10.3390/jof11090685 - 20 Sep 2025
Viewed by 198
Abstract
Non-Saccharomyces yeasts are emerging as promising new probiotics with a beneficial effect equal to or greater than the reference probiotic yeast, Saccharomyces boulardii. Candida intermedia, a non-albicans species not considered a common human pathogen, previously demonstrated probiotic potential. In [...] Read more.
Non-Saccharomyces yeasts are emerging as promising new probiotics with a beneficial effect equal to or greater than the reference probiotic yeast, Saccharomyces boulardii. Candida intermedia, a non-albicans species not considered a common human pathogen, previously demonstrated probiotic potential. In this work, our objective was to evaluate the immunomodulatory effects of C. intermedia ORQ001 in mice vaccinated with inactivated SARS-CoV-2, seeking further evidence of its probiotic activity. Murine macrophages were stimulated with C. intermedia, followed by mRNA transcription analysis via qPCR. Mice were supplemented with C. intermedia prior to SARS-CoV-2 vaccination. Antibody production was assessed by ELISA, and fecal microbiomes were analyzed using next-generation sequencing. C. intermedia significantly increased Il4 and Il13 expression while decreasing Stat3 in macrophages. Splenocytes from supplemented mice exhibited elevated transcription levels of Tnf, Ifng, Il4, Bcl6, and Stat3 after exposure to stimulatory molecules. These mice showed increased levels of anti-SARS-CoV-2 IgG and sIgA isotypes, along with higher abundances of Bacteroides spp. and Clostridium spp. in their gut microbiome. In conclusion, C. intermedia supplementation modulated the expression of key immune-related genes and enhanced humoral responses in mice. Furthermore, its influence on gastrointestinal microbiota suggests a synergistic effect on vaccine immunogenicity. These findings support the potential of C. intermedia as a novel probiotic candidate with immunomodulatory properties applicable to vaccine adjuvanticity. Full article
(This article belongs to the Special Issue Fungal Cell Biology)
Show Figures

Figure 1

20 pages, 1645 KB  
Review
Smart and Functional Probiotic Microorganisms: Emerging Roles in Health-Oriented Fermentation
by Karina Teixeira Magalhães, Raquel Nunes Almeida da Silva, Adriana Silva Borges, Ana Elisa Barbosa Siqueira, Claudia Puerari and Juliana Aparecida Correia Bento
Fermentation 2025, 11(9), 537; https://doi.org/10.3390/fermentation11090537 - 16 Sep 2025
Viewed by 752
Abstract
The incorporation of probiotic microorganisms into fermented foods has long been recognized as a promising strategy to enhance gut health and overall well-being. Conventional probiotics, mainly from the bacterial genera Lactobacillus, Bifidobacterium, Lacticaseibacillus, Levilactobacillus, Lactiplantibacillus and yeast genus Saccharomyces [...] Read more.
The incorporation of probiotic microorganisms into fermented foods has long been recognized as a promising strategy to enhance gut health and overall well-being. Conventional probiotics, mainly from the bacterial genera Lactobacillus, Bifidobacterium, Lacticaseibacillus, Levilactobacillus, Lactiplantibacillus and yeast genus Saccharomyces, contribute to gastrointestinal homeostasis, immune modulation, and metabolic balance. Building on these foundations, recent advances in synthetic biology, systems microbiology, and genetic engineering have enabled the development of smart probiotics: engineered or selectively enhanced strains capable of sensing environmental cues and producing targeted bioactive compounds, such as neurotransmitters and anti-inflammatory peptides. These next-generation microorganisms offer precision functionality in food matrices and hold promise for applications in gastrointestinal health, immune support, and gut–brain axis modulation. However, their deployment also raises critical questions regarding biosafety, regulatory approval, and consumer acceptance. This review provides a comprehensive overview of the mechanisms of action, biotechnological strategies, and health-oriented fermentation applications of smart and functional probiotics, emphasizing their role in the future of personalized and evidence-based functional foods. Full article
Show Figures

Graphical abstract

21 pages, 509 KB  
Review
Microbial Landscapes of the Gut–Biliary Axis: Implications for Benign and Malignant Biliary Tract Diseases
by David Meacci, Angelo Bruni, Alice Cocquio, Giuseppe Dell’Anna, Francesco Vito Mandarino, Giovanni Marasco, Paolo Cecinato, Giovanni Barbara and Rocco Maurizio Zagari
Microorganisms 2025, 13(9), 1980; https://doi.org/10.3390/microorganisms13091980 - 25 Aug 2025
Viewed by 755
Abstract
Next-generation sequencing has overturned the dogma of biliary sterility, revealing low-biomass microbiota along the gut–biliary axis with metabolic and immunologic effects. This review synthesizes evidence on composition, function, and routes of colonization across benign and malignant disease. In cholelithiasis, Proteobacteria- and Firmicutes [...] Read more.
Next-generation sequencing has overturned the dogma of biliary sterility, revealing low-biomass microbiota along the gut–biliary axis with metabolic and immunologic effects. This review synthesizes evidence on composition, function, and routes of colonization across benign and malignant disease. In cholelithiasis, Proteobacteria- and Firmicutes-rich consortia provide β-glucuronidase, phospholipase A2, and bile salt hydrolase, driving bile supersaturation, nucleation, and recurrence. In primary sclerosing cholangitis, primary biliary cholangitis, and autoimmune hepatitis, intestinal dysbiosis and disturbed bile acid pools modulate pattern recognition receptors and bile acid signaling (FXR, TGR5), promote Th17 skewing, and injure cholangiocytes; bile frequently shows Enterococcus expansion linked to taurolithocholic acid. Distinct oncobiomes characterize cholangiocarcinoma subtypes; colibactin-positive Escherichia coli and intratumoral Gammaproteobacteria contribute to DNA damage and chemoresistance. In hepatocellular carcinoma, intratumoral microbial signatures correlate with tumor biology and prognosis. We critically appraise key methodological constraints—sampling route and post-sphincterotomy contamination, antibiotic prophylaxis, low biomass, and heterogeneous analytical pipelines—and outline a translational agenda: validated microbial/metabolomic biomarkers from bile, tissue, and stent biofilms; targeted modulation with selective antibiotics, engineered probiotics, fecal microbiota transplantation, and bile acid receptor modulators. Standardized protocols and spatial, multi-omic prospective studies are required to enable risk stratification and microbiota-informed therapeutics. Full article
(This article belongs to the Special Issue Gut Microbiome in Homeostasis and Disease, 3rd Edition)
Show Figures

Figure 1

34 pages, 1145 KB  
Review
Molecular Mechanisms of Probiotic Action Against Gastrointestinal Cancers
by Christina Thoda and Maria Touraki
Int. J. Mol. Sci. 2025, 26(16), 7857; https://doi.org/10.3390/ijms26167857 - 14 Aug 2025
Viewed by 1760
Abstract
Gastrointestinal (GI) cancers represent a major global health burden. Among them, colorectal cancer (CRC) is the most common type, followed by esophagus, stomach, liver, and pancreatic cancer. Since disturbance of the gut microbiota has been directly associated with the development of severe health [...] Read more.
Gastrointestinal (GI) cancers represent a major global health burden. Among them, colorectal cancer (CRC) is the most common type, followed by esophagus, stomach, liver, and pancreatic cancer. Since disturbance of the gut microbiota has been directly associated with the development of severe health issues, including cancer, probiotic administration may induce dysbiosis reversion and ameliorate carcinogenesis. Therefore, manipulation of the gut microbiota composition based on probiotic utilization has gradually attained scientific interest as a potent therapeutic modality for GI cancers. This review aims to synthesize the current in vitro and in vivo evidence on probiotics’ effectiveness in GI cancer chemoprevention and treatment. It also provides a classification of the fundamental anticancer features of probiotics, including antiproliferation and cell death induction, anticarcinogenic compound production, reduction in chemotherapy-related toxicity, gut microbiota modulation, intestinal barrier improvement, antioxidant activity, immunomodulatory/anti-inflammatory effects, and carcinogen detoxification. Finally, it underscores the future perspectives and challenges of probiotic administration to individuals. In this regard, it emphasizes the exploitation of advanced encapsulation techniques and the development of novel genetically engineered probiotics and next-generation probiotics as feasible ways to improve their bioavailability, ensure their targeted delivery, and eliminate their mild side effects to the host’s health. Full article
(This article belongs to the Special Issue Molecular Advances and Novel Biomarkers in Gastrointestinal Cancers)
Show Figures

Figure 1

26 pages, 1638 KB  
Review
In Silico Modeling of Metabolic Pathways in Probiotic Microorganisms for Functional Food Biotechnology
by Baiken B. Baimakhanova, Amankeldi K. Sadanov, Irina A. Ratnikova, Gul B. Baimakhanova, Saltanat E. Orasymbet, Aigul A. Amitova, Gulzat S. Aitkaliyeva and Ardak B. Kakimova
Fermentation 2025, 11(8), 458; https://doi.org/10.3390/fermentation11080458 - 7 Aug 2025
Cited by 1 | Viewed by 1253
Abstract
Recent advances in computational biology have provided powerful tools for analyzing, modeling, and optimizing probiotic microorganisms, thereby supporting their development as promising agents for improving human health. The essential role of the microbiota in regulating physiological processes and preventing disease has driven interest [...] Read more.
Recent advances in computational biology have provided powerful tools for analyzing, modeling, and optimizing probiotic microorganisms, thereby supporting their development as promising agents for improving human health. The essential role of the microbiota in regulating physiological processes and preventing disease has driven interest in the rational design of next-generation probiotics. This review highlights progress in in silico approaches for enhancing the functionality of probiotic strains. Particular attention is given to genome-scale metabolic models, advanced simulation algorithms, and AI-driven tools that provide deeper insight into microbial metabolism and enable precise probiotic optimization. The integration of these methods with multi-omics data has greatly improved our ability to predict strain behavior and design probiotics with specific health benefits. Special focus is placed on modeling probiotic–prebiotic interactions and host–microbiome dynamics, which are essential for the development of functional food products. Despite these achievements, key challenges remain, including limited model accuracy, difficulties in simulating complex host–microbe systems, and the absence of unified standards for validating in silico-optimized strains. Addressing these gaps requires the development of integrative modeling platforms and clear regulatory frameworks. This review provides a critical overview of current advances, identifies existing barriers, and outlines future directions for the application of computational strategies in probiotic research. Full article
(This article belongs to the Section Probiotic Strains and Fermentation)
Show Figures

Figure 1

20 pages, 1743 KB  
Article
Encapsulation of Lactobacillus reuteri in Chia–Alginate Hydrogels for Whey-Based Functional Powders
by Alma Yadira Cid-Córdoba, Georgina Calderón-Domínguez, María de Jesús Perea-Flores, Alberto Peña-Barrientos, Fátima Sarahi Serrano-Villa, Rigoberto Barrios-Francisco, Marcela González-Vázquez and Minerva Rentería-Ortega
Gels 2025, 11(8), 613; https://doi.org/10.3390/gels11080613 - 4 Aug 2025
Viewed by 1080
Abstract
This study aimed to develop a functional powder using whey and milk matrices, leveraging the protective capacity of chia–alginate hydrogels and the advantages of electrohydrodynamic spraying (EHDA), a non-thermal technique suitable for encapsulating probiotic cells under stress conditions commonly encountered in food processing. [...] Read more.
This study aimed to develop a functional powder using whey and milk matrices, leveraging the protective capacity of chia–alginate hydrogels and the advantages of electrohydrodynamic spraying (EHDA), a non-thermal technique suitable for encapsulating probiotic cells under stress conditions commonly encountered in food processing. A hydrogel matrix composed of chia seed mucilage and sodium alginate was used to form a biopolymeric network that protected probiotic cells during processing. The encapsulation efficiency reached 99.0 ± 0.01%, and bacterial viability remained above 9.9 log10 CFU/mL after lyophilization, demonstrating the excellent protective capacity of the hydrogel matrix. Microstructural analysis using confocal laser scanning microscopy (CLSM) revealed well-retained cell morphology and homogeneous distribution within the hydrogel matrix while, in contrast, scanning electron microscopy (SEM) showed spherical, porous microcapsules with distinct surface characteristics influenced by the encapsulation method. Encapsulates were incorporated into beverages flavored with red fruits and pear and subsequently freeze-dried. The resulting powders were analyzed for moisture, protein, lipids, carbohydrates, fiber, and color determinations. The results were statistically analyzed using ANOVA and response surface methodology, highlighting the impact of ingredient ratios on nutritional composition. Raman spectroscopy identified molecular features associated with casein, lactose, pectins, anthocyanins, and other functional compounds, confirming the contribution of both matrix and encapsulants maintaining the structural characteristics of the product. The presence of antioxidant bands supported the functional potential of the powder formulations. Chia–alginate hydrogels effectively encapsulated L. reuteri, maintaining cell viability and enabling their incorporation into freeze-dried beverage powders. This approach offers a promising strategy for the development of next-generation functional food gels with enhanced probiotic stability, nutritional properties, and potential application in health-promoting dairy systems. Full article
(This article belongs to the Special Issue Food Gels: Fabrication, Characterization, and Application)
Show Figures

Graphical abstract

29 pages, 6122 KB  
Article
Lacticaseibacillus paracasei L21 and Its Postbiotics Ameliorate Ulcerative Colitis Through Gut Microbiota Modulation, Intestinal Barrier Restoration, and HIF1α/AhR-IL-22 Axis Activation: Combined In Vitro and In Vivo Evidence
by Jingru Chen, Linfang Zhang, Yuehua Jiao, Xuan Lu, Ning Zhang, Xinyi Li, Suo Zheng, Bailiang Li, Fei Liu and Peng Zuo
Nutrients 2025, 17(15), 2537; https://doi.org/10.3390/nu17152537 - 1 Aug 2025
Cited by 1 | Viewed by 1294
Abstract
Background: Ulcerative colitis (UC), characterized by chronic intestinal inflammation, epithelial barrier dysfunction, and immune imbalance demands novel ameliorative strategies beyond conventional approaches. Methods: In this study, the probiotic properties of Lactobacillus paracaseiL21 (L. paracaseiL21) and its ability to ameliorate [...] Read more.
Background: Ulcerative colitis (UC), characterized by chronic intestinal inflammation, epithelial barrier dysfunction, and immune imbalance demands novel ameliorative strategies beyond conventional approaches. Methods: In this study, the probiotic properties of Lactobacillus paracaseiL21 (L. paracaseiL21) and its ability to ameliorate colitis were evaluated using an in vitro lipopolysaccharide (LPS)-induced intestinal crypt epithelial cell (IEC-6) model and an in vivo dextran sulfate sodium (DSS)-induced UC mouse model. Results: In vitro, L. paracaseiL21 decreased levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-8) while increasing anti-inflammatory IL-10 levels (p < 0.05) in LPS-induced IEC-6 cells, significantly enhancing the expression of tight junction proteins (ZO-1, occludin, claudin-1), thereby restoring the intestinal barrier. In vivo, both viable L. paracaseiL21 and its heat-inactivated postbiotic (H-L21) mitigated weight loss, colon shortening, and disease activity indices, concurrently reducing serum LPS and proinflammatory mediators. Interventions inhibited NF-κB signaling while activating HIF1α/AhR pathways, increasing IL-22 and mucin MUC2 to restore goblet cell populations. Gut microbiota analysis showed that both interventions increased the abundance of beneficial gut bacteria (Lactobacillus, Dubococcus, and Akkermansia) and improved faecal propanoic acid and butyric acid levels. H-L21 uniquely exerted an anti-inflammatory effect, marked by the regulation of Dubosiella, while L. paracaseiL21 marked by the Akkermansia. Conclusions: These results highlight the potential of L. paracaseiL21 as a candidate for the development of both probiotic and postbiotic formulations. It is expected to provide a theoretical basis for the management of UC and to drive the development of the next generation of UC therapies. Full article
(This article belongs to the Special Issue Probiotics, Postbiotics, Gut Microbiota and Gastrointestinal Health)
Show Figures

Figure 1

11 pages, 1139 KB  
Article
Effect of Akkermansia muciniphila on GLP-1 and Insulin Secretion
by Ananta Prasad Arukha, Subhendu Nayak and Durga Madhab Swain
Nutrients 2025, 17(15), 2516; https://doi.org/10.3390/nu17152516 - 31 Jul 2025
Viewed by 2377
Abstract
Background/Objectives: Gut microbiota research has gained momentum in recent years broadening knowledge of microbial components and their potential effects on health and well-being. Strong association between explicit microbes and metabolic diseases associated with obesity and type 2 diabetes mellitus, gastrointestinal disorders, neurodegenerative diseases, [...] Read more.
Background/Objectives: Gut microbiota research has gained momentum in recent years broadening knowledge of microbial components and their potential effects on health and well-being. Strong association between explicit microbes and metabolic diseases associated with obesity and type 2 diabetes mellitus, gastrointestinal disorders, neurodegenerative diseases, and even cancers have been established. Akkermansia muciniphila is a budding next-generation probiotic that plays an important role in systemic metabolism, intestinal health, and immune regulation, establishing strong implications for its use as a potent therapeutic intervention in diverse diseases. This project aimed at evaluating whether bacterial cell extracts of VH Akkermansia muciniphila (Vidya Strain; VS) can stimulate insulin secretion in INS-1 pancreatic beta cells and GLP-1 secretion in NCI-H716 human L-cells, both established in vitro models for studying metabolic regulation. Methods: Cultured VH Akkermansia muciniphila extracts were administered in a dose-dependent manner on INS-1 cells, and glucose-stimulated insulin secretion (GSIS) was measured via ELISA. Treated Human L-cell lines (NCI-H716) were analyzed for GLP-1 secretion. Results: Our study demonstrated that VH Akkermansia muciniphila extracts modestly increase insulin secretion from INS-1 beta cells and, more notably, induce a robust, dose-dependent rise in GLP-1 secretion from NCI-H716 L-cells, with the highest dose achieving over a 2000% increase comparable to glutamine. Conclusions: These findings suggest that VH A. muciniphila extracts may offer metabolic benefits by enhancing GLP-1 release, highlighting their potential for managing type 2 diabetes and obesity. Full article
(This article belongs to the Section Prebiotics and Probiotics)
Show Figures

Figure 1

17 pages, 4949 KB  
Article
Apple Juice Fermented with Lactiplantibacillus plantarum Improves Its Flavor Profile and Probiotic Potential
by Boqian Zhou, Zhuobin Xing, Yiting Wang, Xin Guan, Fuyi Wang, Jiaqi Yin, Zhibo Li, Qiancheng Zhao, Hongman Hou and Xue Sang
Foods 2025, 14(13), 2373; https://doi.org/10.3390/foods14132373 - 4 Jul 2025
Viewed by 862
Abstract
Fermented apple juice (FAJ), a nutrient-dense beverage rich in vitamins, offers multiple health benefits, including improved digestion, enhanced fat metabolism, and sustained energy provision with reduced caloric intake. To advance the development of probiotic-enriched flavored and functional juices, this study establishes Lactiplantibacillus plantarum [...] Read more.
Fermented apple juice (FAJ), a nutrient-dense beverage rich in vitamins, offers multiple health benefits, including improved digestion, enhanced fat metabolism, and sustained energy provision with reduced caloric intake. To advance the development of probiotic-enriched flavored and functional juices, this study establishes Lactiplantibacillus plantarum (L. plantarum) as a safe and effective starter culture for apple juice fermentation. The selected strain exhibited minimal biogenic amine synthesis, producing only 30.55 ± 1.2 mg/L of putrescine and 0.59 ± 0.55 mg/L of cadaverine, while histamine and tyramine were undetectable. Furthermore, the strain demonstrated no hemolytic activity and exhibited robust biofilm-forming capacity, reinforcing its suitability for fermentation applications. An electronic nose analysis revealed that L. plantarum significantly enriched the volatile compound profile of FAJ, leading to an improved flavor profile. The strain also displayed excellent growth adaptability in the apple juice matrix, further optimizing fermentation efficiency and sensory quality. Crucially, 16S rRNA sequencing demonstrated that FAJ specifically restructures the gut microbiota in obese individuals, significantly elevating the relative abundance of beneficial genera, including Enterococcus, Parabacteroides, and Bifidobacterium (p < 0.05). Concurrently, FAJ enhanced glycolytic activity, suggesting a potential role in metabolic regulation. Collectively, these findings confirm that L. plantarum-fermented FAJ combines favorable sensory properties and safety with promising anti-obesity effects mediated through gut microbiome modulation and metabolic pathway activation. This study provides a critical scientific foundation for designing next-generation functional fermented beverages with targeted health benefits. Full article
Show Figures

Figure 1

11 pages, 3786 KB  
Article
Survival and Morphological Changes of Clostridium butyricum Spores Co-Exposed to Antibiotics and Simulated Gastrointestinal Fluids: Implications for Antibiotic Stewardship
by Yi-Meng Yang, Meng-Yue Zhang, Ying-Ying Wu, Lu Zhang and Yi-Xuan Zhang
Microorganisms 2025, 13(6), 1347; https://doi.org/10.3390/microorganisms13061347 - 10 Jun 2025
Viewed by 1188
Abstract
Probiotics are often advised to be taken separately from antibiotics due to their sensitivity to antibiotic activity and gastrointestinal (GI) stress. However, Clostridium butyricum spores, as next-generation probiotics, may withstand concurrent use with antibiotics due to their unique structural adaptations. This study systematically [...] Read more.
Probiotics are often advised to be taken separately from antibiotics due to their sensitivity to antibiotic activity and gastrointestinal (GI) stress. However, Clostridium butyricum spores, as next-generation probiotics, may withstand concurrent use with antibiotics due to their unique structural adaptations. This study systematically evaluated the survival rates and morphological changes of C. butyricum spores exposed to 10 clinically relevant antibiotics in simulated gastric/intestinal fluids, exploring their feasibility for co-administration with antibiotics. Survival rates of C. butyricum spores were tested against 10 antibiotics across four classes (β-lactams, macrolides, aminoglycosides, and tetracyclines) in simulated GI fluids. Time–kill curves analyzed spore survival over 0–4 h, while scanning electron microscopy (SEM) observed spore wall integrity and morphological changes under different conditions. The spore survival rates remained >89% in intestinal fluid and >60% in gastric fluid across all antibiotics. SEM revealed gastric acid and proteolysis increased spore wall fragmentation, reducing resistance, whereas the intestinal environment preserved spore integrity. This study was the first to demonstrate that C. butyricum spores can survive simultaneous exposure to antibiotics in the gastrointestinal tract, challenging traditional probiotic usage guidelines. The findings support their co-administration with antibiotics to simplify dosing regimens and improve medication adherence. Such an approach advances antimicrobial stewardship by optimizing therapeutic strategies for antibiotic–probiotic combinations. Full article
(This article belongs to the Section Antimicrobial Agents and Resistance)
Show Figures

Figure 1

24 pages, 1816 KB  
Review
A Systematic Review on Microbial Profiling Techniques in Goat Milk: Implications for Probiotics and Shelf-Life
by Nare Jessica Monareng, Keabetswe T. Ncube, Charles van Rooi, Mamokoma C. Modiba and Bohani Mtileni
Int. J. Mol. Sci. 2025, 26(12), 5551; https://doi.org/10.3390/ijms26125551 - 10 Jun 2025
Viewed by 1352
Abstract
Due to its high digestibility, rich nutrient profile, and potential probiotic content, goat milk is an essential nutritional resource, particularly for individuals with cow milk allergies. This review summarises the current state of microbial diversity in goat milk, emphasising the implications for quality, [...] Read more.
Due to its high digestibility, rich nutrient profile, and potential probiotic content, goat milk is an essential nutritional resource, particularly for individuals with cow milk allergies. This review summarises the current state of microbial diversity in goat milk, emphasising the implications for quality, safety, and probiotic potential. This systematic review adhered to PRISMA guidelines, conducting a comprehensive literature search across PubMed, ScienceDirect, and Google Scholar using keywords related to microbial profiling in goat milk. The inclusion criteria targeted English-language studies from 2000 to 2025 that utilised high-throughput or next-generation sequencing methods. Out of 126 articles screened, 84 met the eligibility criteria. The extracted data focused on microbial diversity, profiling techniques, and their respective strengths and limitations in evaluating probiotic potential and spoilage risks. The review addresses the challenges linked to microbial spoilage and the composition and functional roles of microbial communities in goat milk. With species such as Bacillus and Pseudomonas playing crucial roles in fermentation and spoilage, key findings emphasise the prevalence of microbial phyla, including Proteobacteria, Firmicutes, and Actinobacteria in goat milk. The review also explores the probiotic potential of the goat milk microbiota, highlighting the health benefits associated with strains such as Lactobacillus and Bifidobacterium. Significant discoveries underline the necessity for advanced multi-omics techniques to thoroughly define microbial ecosystems and the substantial gaps in breed-specific microbiota research. Important findings illustrate the need for enhanced multi-omics techniques, given the challenges of host RNA and protein interference, low microbial biomass, and limited goat-specific reference databases, for optimising probiotic development, spoilage prevention strategies, and integrating metagenomics, metabolomics, metaproteomics, and metatranscriptomics to improve milk quality and safety as some of the future research objectives. This study emphasises the importance of understanding goat milk microbiology to advance dairy science and enhance human health. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Back to TopTop