Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (68)

Search Parameters:
Keywords = bacteria-derived extracellular vesicles

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4179 KiB  
Article
Camel Milk-Derived Extracellular Vesicles as a Functional Food Component Ameliorate Hypobaric Hypoxia-Induced Colonic Injury Through Microbiota–Metabolite Crosstalk
by Hui Yang, Demtu Er, Yu-Huan Wang, Bin-Tao Zhai and Rili Ge
Nutrients 2025, 17(15), 2431; https://doi.org/10.3390/nu17152431 - 25 Jul 2025
Viewed by 346
Abstract
Background/Objectives: This study investigates the therapeutic potential of camel milk-derived extracellular vesicles (CM-EVs) for treating colonic damage caused by high-altitude hypoxia, supporting the WHO’s “Food as Medicine” initiative. Methods: Using a 5500 m mouse model, researchers induced colonic injury and treated it with [...] Read more.
Background/Objectives: This study investigates the therapeutic potential of camel milk-derived extracellular vesicles (CM-EVs) for treating colonic damage caused by high-altitude hypoxia, supporting the WHO’s “Food as Medicine” initiative. Methods: Using a 5500 m mouse model, researchers induced colonic injury and treated it with oral CM-EVs for 15 days, comparing results to whole camel milk. Results: CM-EVs outperformed whole milk, significantly improving colon health by restoring barrier integrity and reducing disease activity index (DAI) (p < 0.01). They boosted beneficial bacteria like Lactobacillus and Bifidobacterium and decreased Enterobacteriaceae (p < 0.01). Metabolic analysis showed restored bile acid balance and amino acid modulation via the FXR/NF-κB pathway, reducing TLR4/MyD88-mediated inflammation and oxidative stress (p < 0.01). Fecal microbiota transplantation in the CM-EVs group notably decreased DAI and increased colon length (p < 0.05). Conclusions: CM-EVs repair mucosal damage, balance microbiota, and regulate metabolism to combat hypoxia-induced colonic damage, suggesting their potential as nutraceuticals and altitude-adaptive foods. This showcases nanotechnology’s role in enhancing traditional dietary benefits via precision nutrition. Full article
Show Figures

Figure 1

17 pages, 2652 KiB  
Article
Spontaneous Bacterial Peritonitis in Advanced Cirrhosis: Diagnosis by Tm Mapping and Inflammatory Profiles of Extracellular Vesicles
by Aiko Murayama, Kazuto Tajiri, Nozomu Muraishi, Yuka Hayashi, Masami Minemura, Hideki Niimi and Ichiro Yasuda
J. Clin. Med. 2025, 14(14), 5096; https://doi.org/10.3390/jcm14145096 - 17 Jul 2025
Viewed by 274
Abstract
Background/Objectives: Ascites is a major complication in patients with decompensated cirrhosis. Spontaneous bacterial peritonitis (SBP), an infection of the ascitic fluid, is a life-threatening condition in patients with cirrhosis. This study aimed to assess the utility of Tm mapping, a novel high-efficacy [...] Read more.
Background/Objectives: Ascites is a major complication in patients with decompensated cirrhosis. Spontaneous bacterial peritonitis (SBP), an infection of the ascitic fluid, is a life-threatening condition in patients with cirrhosis. This study aimed to assess the utility of Tm mapping, a novel high-efficacy method for bacterial detection and quantification, in the early diagnosis of SBP and its pathogenesis. Methods: Ascitic fluid samples from 29 patients with cirrhosis were analyzed using Tm mapping for bacterial identification. Inflammatory cytokine and pathogen-associated molecular pattern levels in ascitic fluid were measured and correlated with SBP pathophysiology. Additionally, the role of ascitic macrophages was investigated in vitro. Results: Tm mapping detected bacteria more effectively than conventional culture methods. In samples where bacteria were identified, ascitic interleukin (IL)-6 levels were elevated. A positive correlation was observed between extracellular vesicle (EV) levels and IL-6, suggesting a role for EVs in peritoneal inflammation. Furthermore, EVs derived from Gram-negative bacteria induced M1 macrophage differentiation via the signal transducer and activator of transcription 1 signaling pathway. Conclusions: Tm mapping is a valuable tool for the early detection of bacteria in ascitic fluid. Additionally, EVs promote M1 macrophage differentiation, implicating them in the pathogenesis of cirrhotic complications, including SBP. Full article
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)
Show Figures

Figure 1

33 pages, 4665 KiB  
Review
A Paradigm Shift in SSTI Management: The Multifunctional Role of Extracellular Vesicles
by Barathan Muttiah and Alfizah Hanafiah
Int. J. Mol. Sci. 2025, 26(13), 6481; https://doi.org/10.3390/ijms26136481 - 5 Jul 2025
Viewed by 683
Abstract
Skin and soft tissue infections (SSTIs) are becoming an urgent public health issue worldwide. The globe is facing a growing problem with drug-resistant germs, and current treatments are not quite cutting it. There is a real need for new therapies that can tackle [...] Read more.
Skin and soft tissue infections (SSTIs) are becoming an urgent public health issue worldwide. The globe is facing a growing problem with drug-resistant germs, and current treatments are not quite cutting it. There is a real need for new therapies that can tackle these challenges more effectively. This brings us to an interesting question: Can extracellular vesicles (EVs) from different sources, such as mesenchymal stem cells (MSCs), immune cells, or even plants and animals, help in treating SSTIs, especially given the rise in drug resistance? Studies have shown that MSC-derived EVs are particularly noteworthy because they carry components such as antimicrobial peptides (AMPs) that can work together to fight infections, boost the immune response, and aid in healing. These vesicles play a role in how our body interacts with infections, helping to clear bacteria, reduce inflammation, and promote tissue repair. We also see that EVs from plants and bacteria can directly fight off germs, while those from animals can support the healing process of skin. Although early studies have shown promise for EV therapies, there are still hurdles to overcome, such as ensuring consistent production and delivery. This review looks at the potential of EVs as powerful agents in managing infections and supporting healing, highlighting an exciting area of research in medicine. Full article
(This article belongs to the Special Issue Extracellular Vesicles: Advances in Multi-Omics)
Show Figures

Graphical abstract

16 pages, 512 KiB  
Review
The Role of Helicobacter pylori Heat Shock Proteins in Gastric Diseases’ Pathogenesis
by Olga Maria Manna, Celeste Caruso Bavisotto, Melania Ionelia Gratie, Provvidenza Damiani, Giovanni Tomasello and Francesco Cappello
Int. J. Mol. Sci. 2025, 26(11), 5065; https://doi.org/10.3390/ijms26115065 - 24 May 2025
Cited by 1 | Viewed by 1940
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach and is associated with several gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. The bacterium’s ability to thrive in the harsh gastric environment is due, to [...] Read more.
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach and is associated with several gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. The bacterium’s ability to thrive in the harsh gastric environment is due, to some extent, to its stress response mechanisms, with its heat shock proteins (HSPs) playing a putative, yet not fully understood, role in these adaptive processes. HSPs are a family of molecules, highly conserved throughout phylogenesis, that assist in protein folding, prevent aggregation, and ensure cellular homeostasis under stressful conditions. In H. pylori, HSPs contribute to survival in the stomach’s acidic environment and oxidative stress. Furthermore, they aid in the bacterium’s ability to adhere to gastric epithelial cells, modulate the host immune response, and form biofilms, all contributing to chronic infection and pathogenicity. The role of microbial HSPs in antibiotic resistance has also emerged as a critical area of research, as these proteins help stabilize efflux pumps, protect essential proteins targeted by antibiotics, and promote biofilm formation, thereby reducing the efficacy of antimicrobial treatments. Among bacterial HSPs, GroEL and DnaK are probably the major proteins that control most of the H. pylori’s functioning. Indeed, both proteins possess remarkable acid resistance, high substrate affinity, and dual roles in protein homeostasis and host interaction. These features make them critical for H. pylori’s adaptation, persistence, and pathogenicity in the gastric niche. In addition, recent findings have also highlighted the involvement of HSPs in the crosstalk between H. pylori and gastric epithelial cells mediated by the release of bacterial outer membrane vesicles and host-derived exosomes, both of these extracellular vesicles being part of the muco-microbiotic layer of the stomach and influencing cellular signalling and immune modulation. Considering their critical role in the survival and persistence of bacteria, microbial HSPs also represent potential therapeutic targets. Strategies aimed at inhibiting microbial HSP function, combined with conventional antibiotics or developing vaccines targeting microbial HSPs, could provide new avenues for the treatment of H. pylori infections and combat antibiotic resistance. This review explores the multifaceted roles of microbial HSPs in the pathogenesis of H. pylori, highlighting their contributions to bacterial adhesion, immune evasion, stress response, and antibiotic resistance. Full article
(This article belongs to the Special Issue Pathogenicity and Antibiotic Resistance of Helicobacter pylori)
Show Figures

Graphical abstract

26 pages, 9145 KiB  
Article
Benzimidazole-Derived B2 as a Fluorescent Probe for Bacterial Outer Membrane Vesicle (OMV) Labeling: Integrating DFT, Molecular Dynamics, Flow Cytometry, and Confocal Microscopy
by Francisco Parra, Alexander Carreño, Evys Ancede-Gallardo, Diana Majluf, Jorge A. Soto, Romina V. Sepúlveda, Daniel Aguayo, María Carolina Otero, Iván L. Calderón, Fernando Gil and Juan A. Fuentes
Int. J. Mol. Sci. 2025, 26(10), 4682; https://doi.org/10.3390/ijms26104682 - 14 May 2025
Viewed by 965
Abstract
Bacterial outer membrane vesicles (OMVs) are nanoscale extracellular structures produced by Gram-negative bacteria that are critical for microbial biology and host-pathogen interactions and have great potential in biotechnological applications. Despite the availability of fluorescent dyes for OMV studies, many are repurposed from eukaryotic [...] Read more.
Bacterial outer membrane vesicles (OMVs) are nanoscale extracellular structures produced by Gram-negative bacteria that are critical for microbial biology and host-pathogen interactions and have great potential in biotechnological applications. Despite the availability of fluorescent dyes for OMV studies, many are repurposed from eukaryotic extracellular vesicle research and are not explicitly optimized for OMVs, leading to challenges in achieving consistent labeling, minimizing background noise, and preserving vesicle integrity during analyses. This study evaluates B2, a benzimidazole-derived fluorophore, for OMV labeling in advanced techniques like flow cytometry and confocal microscopy. OMVs were isolated from Escherichia coli strains BL21 and O157, and their integrity was confirmed using transmission electron microscopy (TEM). B2 staining protocols were optimized for OMVs, and fluorescence analyses revealed specific interactions with the vesicle membranes, reducing aggregation and enhancing signal uniformity. Flow cytometry indicated near-complete labeling efficiency (98–100%) with minimal background interference. Confocal microscopy further validated B2’s effectiveness, showing evident OMV internalization into epithelial HT-29 cells and compatibility with other fluorophores. Density functional theory (DFT) calculations, including Fukui function analysis, identified key electrophilic and nucleophilic regions in B2 that facilitate specific hydrogen bonding and polar interactions with membrane components. Non-covalent interaction (NCI) analysis revealed pronounced intramolecular hydrogen bonding along with discrete regions of weak van der Waals interactions. Molecular dynamics simulations suggest that B2 exhibits an affinity for both the hydrophobic core of the lipid bilayer and the core oligosaccharide region of the LPS layer, which collectively ensures sustained retention of the dye. The findings presented in this study position B2 as a valuable fluorophore for OMV research. Full article
Show Figures

Graphical abstract

20 pages, 4427 KiB  
Article
Separation and Characterization of Heterogeneity Among Various Sizes of Outer Membrane Vesicles Derived from the Probiotic Escherichia coli Nissle 1917
by Ning Li, Hongbo Xin and Keyu Deng
Membranes 2025, 15(5), 141; https://doi.org/10.3390/membranes15050141 - 5 May 2025
Cited by 1 | Viewed by 1006
Abstract
Outer membrane vesicles (OMVs) are extracellular vesicles secreted by Gram-negative bacteria with diameters of 20–250 nm. OMVs contain various biologically active substances from their parent bacteria, such as proteins, lipids, and nucleic acids. Escherichia coli Nissle 1917 (EcN) is a Gram-negative probiotic that [...] Read more.
Outer membrane vesicles (OMVs) are extracellular vesicles secreted by Gram-negative bacteria with diameters of 20–250 nm. OMVs contain various biologically active substances from their parent bacteria, such as proteins, lipids, and nucleic acids. Escherichia coli Nissle 1917 (EcN) is a Gram-negative probiotic that resides in the human intestine. EcN-derived OMVs are pivotal in modulating intestinal immune responses. However, few studies have addressed the heterogeneity of EcN-derived OMVs in terms of size, significantly limiting the research on their clinical applications. Currently, there are a lack of feasible methods for obtaining EcN-derived OMVs of different sizes. To address this knowledge gap, we developed a membrane filtration method to isolate EcN-derived OMVs of varying sizes. In this study, we first used gradient filtration to isolate high-purity EcN-derived OMVs and conducted a proteomic analysis. Subsequently, we used membrane filtration to separate the EcN-derived OMVs by size. We successfully obtained EcN-derived OMVs of three specific sizes: <50 nm, 50–100 nm, and 100–300 nm. We then performed proteomic analyses of these EcN-derived OMVs and compared their protein profiles. Finally, we compared the ability of each EcN-derived OMV type to induce RAW264.7 macrophages to secrete the pro-inflammatory factor interleukin (IL)-1β and the anti-inflammatory factor IL-10. The EcN-derived OMVs contained 646 different proteins overall; those of different sizes contained different protein types. Among them, the EcN-derived OMVs in the <50 nm group contained significantly fewer proteins (262 different types in total) than those in the 50–100 nm (1603 types) and 100–300 nm (1568 types) groups. Furthermore, the <50 nm group had fewer membrane proteins (40) than the 50–100 nm (215) and 100–300 nm (209) groups. We also found that RAW264.7 macrophages secreted different concentrations of IL-1β and IL-10 following co-incubation with the three EcN-derived OMV types. The 50–100 nm EcN-derived OMV group showed a stronger effect in terms of inducing inflammatory cytokine secretion compared to the other two groups. This study provides direct experimental evidence that EcN-derived OMVs of different sizes exhibit heterogeneous properties. Full article
Show Figures

Figure 1

28 pages, 2126 KiB  
Review
Snake Venom Compounds: A New Frontier in the Battle Against Antibiotic-Resistant Infections
by Barathan Muttiah and Alfizah Hanafiah
Toxins 2025, 17(5), 221; https://doi.org/10.3390/toxins17050221 - 1 May 2025
Viewed by 1680
Abstract
The occurrence of antibiotic-resistant bacteria is a serious global health issue, and it emphasizes the need for novel antimicrobial agents. This review explores the potential of snake venom as another alternative strategy against antimicrobial resistance. Snake venoms are complex combinations of bioactive peptides [...] Read more.
The occurrence of antibiotic-resistant bacteria is a serious global health issue, and it emphasizes the need for novel antimicrobial agents. This review explores the potential of snake venom as another alternative strategy against antimicrobial resistance. Snake venoms are complex combinations of bioactive peptides and proteins, including metalloproteases (MPs), serine proteases (SPs), phospholipase A2 (PLA2) enzymes, three-finger toxins (3FTXs), cysteine-rich secretory proteins (CRISPs), L-amino acid oxidases (LAAOs), and antimicrobial peptides (AMPs). The antibacterial products possess wide-spectrum antibacterial activity against resistant microbes via diverse mechanisms such as cell membrane disruption, enzymatic hydrolysis of microbial structures, generation of oxidative stress, inhibition of biofilms, and immunomodulation. Strong antimicrobial activity is reported by most studies, but these are mostly restricted to in vitro testing with low translational use. Although preliminary insights into molecular targets and physiological effects exist, further studies are needed to clarify long-term safety and therapeutic potential. Special attention is given to snake venom-derived extracellular vesicles (SVEVs), which enhance the therapeutic potential of venom toxins by protecting them from degradation, improving bioavailability, and facilitating targeted delivery. Furthermore, innovative delivery strategies such as PEGylation, liposomes, hydrogels, microneedle patches, biopolymer films, and nanoparticles are discussed for their role in reducing systemic toxicity and enhancing antimicrobial efficacy. The rational modification of venom-derived peptides further expands their therapeutic utility by improving pharmacokinetics and minimizing off-target effects. Together, these approaches highlight the translational potential of snake venom-based therapies as next-generation antimicrobials in the fight against resistant infections. By outlining these challenges and directions, this review positions snake venom as an overlooked but fertile resource in the battle against antibiotic resistance. Full article
Show Figures

Figure 1

18 pages, 5328 KiB  
Article
Rigorous Process for Isolation of Gut-Derived Extracellular Vesicles (EVs) and the Effect on Latent HIV
by Nneoma C. J. Anyanwu, Lakmini S. Premadasa, Wasifa Naushad, Bryson C. Okeoma, Mahesh Mohan and Chioma M. Okeoma
Cells 2025, 14(8), 568; https://doi.org/10.3390/cells14080568 - 9 Apr 2025
Viewed by 766
Abstract
The human gastrointestinal (GI) track host trillions of microorganisms that secrete molecules, including extracellular vesicles (EVs) and extracellular condensates (ECs) that may affect physiological and patho-physiological activities in the host. However, efficient protocols for the isolation of pure and functional GI-derived EVs|ECs is [...] Read more.
The human gastrointestinal (GI) track host trillions of microorganisms that secrete molecules, including extracellular vesicles (EVs) and extracellular condensates (ECs) that may affect physiological and patho-physiological activities in the host. However, efficient protocols for the isolation of pure and functional GI-derived EVs|ECs is lacking. Here, we describe the use of high-resolution particle purification liquid chromatography (PPLC) gradient-bead-column integrated with polyvinylpolypyrrolidone (PVPP)-mediated extraction of impurities to isolate EVs from colonic content (ColEVs). PVPP facilitates the isolation of pure, non-toxic, and functionally active ColEVs that were internalized by cells and functionally activate HIV LTR promoter. ColEVs isolated without PVPP have a reductive effect on MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) without living cells, suggesting that ColEVs contain reductases capable of catalyzing the reduction of MTT to formazan. The assessment of the origin of ColEVs reveals that they are composed of both bacteria and host particles. This protocol requires ~12 h (5 h preprocessing, 7 h isolation) to complete and should be used to purify EVs from sources contaminated with microbial agents to improve rigor. This protocol provides a robust tool for researchers and clinicians investigating GI-derived EVs and the translational use of GI-derived EVs for diagnostic and therapeutic use. Additionally, GI-derived EVs may serve as a window into the pathogenesis of diseases. Full article
Show Figures

Figure 1

15 pages, 857 KiB  
Review
Innate Immunity in Cystic Fibrosis: Varied Effects of CFTR Modulator Therapy on Cell-to-Cell Communication
by Jennifer Hynes, Clifford C. Taggart, Rabindra Tirouvanziam and Judith A. Coppinger
Int. J. Mol. Sci. 2025, 26(6), 2636; https://doi.org/10.3390/ijms26062636 - 14 Mar 2025
Cited by 1 | Viewed by 1164
Abstract
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to [...] Read more.
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to eventual respiratory failure. Bidirectional crosstalk between epithelial cells, leukocytes (e.g., neutrophils, macrophages) and bacteria via release of intra-cellular mediators is key to driving inflammation in CF airways. In recent years, a highly effective combination of therapeutics targeting the CFTR defect have revolutionized treatment in CF. Despite these advancements and due to the complexity of the immune response in the CF airway, the full impact of highly effective modulator therapy (HEMT) on airway inflammation is not fully determined. This review provides the evidence to date on crosstalk mechanisms between host epithelium, leukocytes and bacteria and examines the effect of HEMT on both soluble and membrane-derived immune mediators in clinical samples. The varied effects of HEMT on expression of key proteases, cytokines and extracellular vesicles (EVs) in relation to clinical parameters is assessed. Advances in treatment with HEMT have shown potential in dampening the chronic inflammatory response in CF airways. However, to fully quell inflammation and maximize lung tissue resilience, further interventions may be necessary. Exploring the effects of HEMT on key immune mediators paves the way for identifying new anti-inflammatory approaches targeting host immune cell interactions, such as EV-directed lung therapies. Full article
(This article belongs to the Special Issue New Research Insights in Cystic Fibrosis and CFTR-Related Diseases)
Show Figures

Figure 1

19 pages, 2387 KiB  
Review
Extracellular Vesicles in Bacteria, Archaea, and Eukaryotes: Mechanisms of Inter-Kingdom Communication and Clinical Implications
by Maria Di Naro, Giulio Petronio Petronio, Farwa Mukhtar, Marco Alfio Cutuli, Irene Magnifico, Marilina Falcone, Natasha Brancazio, Antonio Guarnieri, Roberto Di Marco and Daria Nicolosi
Microorganisms 2025, 13(3), 636; https://doi.org/10.3390/microorganisms13030636 - 11 Mar 2025
Viewed by 1685
Abstract
Living organisms must adapt and communicate effectively in their environment to survive. Cells communicate through various mechanisms, including releasing growth factors, chemokines, small bioactive molecules, and cell–cell contact. In recent years, a new and sophisticated cell communication mechanism based on extracellular vesicles (EVs) [...] Read more.
Living organisms must adapt and communicate effectively in their environment to survive. Cells communicate through various mechanisms, including releasing growth factors, chemokines, small bioactive molecules, and cell–cell contact. In recent years, a new and sophisticated cell communication mechanism based on extracellular vesicles (EVs) has been described in all three domains of life: archaea, bacteria, and eukaryotes. EVs are small, bilayer proteolipid vesicles released by cells into the extracellular space. This review aims to analyze and compare the current literature on bacterial, archaeal, and eukaryotic EVs and their possible clinical applications. This framework will address three key points: (a) The role of EVs in bacteria, eukaryotes, and archaea. (b) What is the impact of EVs in archaea on disease? (c) How archaea use EVs to communicate with other domains (bacteria/eukaryotes). Full article
(This article belongs to the Collection Feature Papers in Medical Microbiology)
Show Figures

Figure 1

15 pages, 3970 KiB  
Article
Targeting Inflammation and Skin Aging via the Gut–Skin Axis: The Role of Lactiplantibacillus plantarum HY7714-Derived Extracellular Vesicles
by Hayera Lee, Yun-Ha Lee, Dong-Ki Hong, Sung-Jun Mo, Soomin Jeon, Soo-Dong Park, Jae-Jung Shim, Jeong-Lyoul Lee and Jae-Hwan Lee
Microorganisms 2024, 12(12), 2466; https://doi.org/10.3390/microorganisms12122466 - 30 Nov 2024
Cited by 1 | Viewed by 2285
Abstract
Intestinal mucosal tissues are prone to infections, often leading to inflammation. Lactic acid bacteria in the gut can modulate these inflammatory responses, but the interaction between host cells and lactic acid bacteria remains unclear. This study examines how Lactiplantibacillus plantarum HY7714 alleviates intestinal [...] Read more.
Intestinal mucosal tissues are prone to infections, often leading to inflammation. Lactic acid bacteria in the gut can modulate these inflammatory responses, but the interaction between host cells and lactic acid bacteria remains unclear. This study examines how Lactiplantibacillus plantarum HY7714 alleviates intestinal inflammation using gut-on-a-chip technology and in vitro models. Inflammation was induced using a gut-on-a-chip, and changes in cell morphology and barrier function were analyzed. Extracellular vesicles (EVs) derived from HY7714-improved intestinal cell structure repaired damage and restored tight junction integrity. Additionally, they attenuated inflammatory cytokines by regulating the MyD88/mTOR/NF-κB signaling pathway. RNA sequencing revealed downregulation of vicinal oxygen chelate (VOC) family proteins and proline aminopeptidase, both linked to inflammation and extracellular matrix interactions in skin health. Therefore, we explored the effects of HY7714 EVs on skin cells. The findings showed that HY7714 EVs reduced cytotoxicity and downregulated metalloproteinase expression in skin cells exposed to UVB radiation, indicating their potential anti-aging and anti-photoaging properties. These findings suggest that HY7714-derived EVs enhance both intestinal and skin health by reducing inflammation and improving barrier function, with potential benefits for the gut–skin axis. Full article
(This article belongs to the Special Issue Probiotic and Postbiotic Properties of Lactobacillus)
Show Figures

Figure 1

21 pages, 6051 KiB  
Systematic Review
The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies
by Leila Khalili, Gwoncheol Park, Ravinder Nagpal and Gloria Salazar
Microorganisms 2024, 12(8), 1627; https://doi.org/10.3390/microorganisms12081627 - 9 Aug 2024
Cited by 12 | Viewed by 8286
Abstract
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila’s action and its effects on the microbiome are not well [...] Read more.
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila’s action and its effects on the microbiome are not well understood. In this study, we examined the influence of A. muciniphila and its derivatives on gastrointestinal (GI) and metabolic disorders through a meta-analysis of studies conducted on mouse models. A total of 39 eligible studies were identified through targeted searches on PubMed, Web of Science, Science Direct, and Embase until May 2024. A. muciniphila (alive or heat-killed) and its derivatives positively affected systemic and gut inflammation, liver enzyme level, glycemic response, and lipid profiles. The intervention increased the expression of tight-junction proteins in the gut, improving gut permeability in mouse models of GI and metabolic disorders. Regarding body weight, A. muciniphila and its derivatives prevented weight loss in animals with GI disorders while reducing body weight in mice with metabolic disorders. Sub-group analysis indicated that live bacteria had a more substantial effect on most analyzed biomarkers. Gut microbiome analysis using live A. muciniphila identified a co-occurrence cluster, including Desulfovibrio, Family XIII AD3011 group, and Candidatus Saccharimonas. Thus, enhancing the intestinal abundance of A. muciniphila and its gut microbial clusters may provide more robust health benefits for cardiometabolic, and age-related diseases compared with A. muciniphila alone. The mechanistic insight elucidated here will pave the way for further exploration and potential translational applications in human health. Full article
(This article belongs to the Special Issue Advances in Host-Gut Microbiota)
Show Figures

Figure 1

14 pages, 1603 KiB  
Article
Fermented Cultured Wild Ginseng Roots (Panax ginseng C.A. Meyer) Using Limosilactobacillus fermentum HY7303 Enhances the Intestinal Barrier by Bioconversion of Ginsenosides and Extracellular Vesicle Production
by Sung-Joon Mo, Eun-Ji Kim, Yun-Ha Lee, Soo-Dong Park, Jae-Jung Shim, Jung-Lyul Lee and Jae-Hwan Lee
Fermentation 2024, 10(7), 362; https://doi.org/10.3390/fermentation10070362 - 17 Jul 2024
Cited by 1 | Viewed by 1723
Abstract
Wild ginseng is known to have better pharmacological effects than cultivated ginseng. Additionally, recently developed bioengineering technology has made it possible to produce cultured wild ginseng with the same genetic composition. In this study, we investigated the change in characteristics and the improvement [...] Read more.
Wild ginseng is known to have better pharmacological effects than cultivated ginseng. Additionally, recently developed bioengineering technology has made it possible to produce cultured wild ginseng with the same genetic composition. In this study, we investigated the change in characteristics and the improvement of the intestinal barrier of cultured wild ginseng roots (CWG) and fermented cultured wild ginseng roots (FCWG). First, we screened nine strains of bacteria that are capable of growing on 5-brix CWG medium, and Limosilactobacillus fermentum HY7303 (HY7303) showed the highest growth. Second, changes in the characteristics of CWG due to fermentation using HY7303 showed that pH and total carbohydrates decreased, and reducing sugars increased. The contents of minor ginsenosides (Rg3(s), Rk1, and Rg5) increased. Third, extracellular vesicles (EVs) with a single peak at 493.7 nm were isolated from CWG, and EVs with three peaks at 9.0 nm, 155.6 nm, and 459.0 nm were isolated from FCWG, respectively. Finally, when we treated Caco-2 cells with FCWG and EVs, we confirmed the improvement of intestinal barrier functions, including recovery, permeability, and expression of tight-junction protein genes. In this study, we confirmed the potential pharmacological effects of minor ginsenosides and EVs derived from FCWG. In conclusion, this study suggests that CWG fermentation with HY7303 improves the intestinal barrier by increasing minor ginsenosides and producing EVs. Full article
Show Figures

Figure 1

38 pages, 4080 KiB  
Review
Postbiotics as Molecules Targeting Cellular Events of Aging Brain—The Role in Pathogenesis, Prophylaxis and Treatment of Neurodegenerative Diseases
by Pola Głowacka, Katarzyna Oszajca, Agnieszka Pudlarz, Janusz Szemraj and Monika Witusik-Perkowska
Nutrients 2024, 16(14), 2244; https://doi.org/10.3390/nu16142244 - 12 Jul 2024
Cited by 18 | Viewed by 5156
Abstract
Aging is the most prominent risk factor for neurodegeneration occurrence. The most common neurodegenerative diseases (NDs), Alzheimer’s (AD) and Parkinson’s (PD) diseases, are characterized by the incidence of proteinopathy, abnormal activation of glial cells, oxidative stress, neuroinflammation, impaired autophagy and cellular senescence excessive [...] Read more.
Aging is the most prominent risk factor for neurodegeneration occurrence. The most common neurodegenerative diseases (NDs), Alzheimer’s (AD) and Parkinson’s (PD) diseases, are characterized by the incidence of proteinopathy, abnormal activation of glial cells, oxidative stress, neuroinflammation, impaired autophagy and cellular senescence excessive for the patient’s age. Moreover, mitochondrial disfunction, epigenetic alterations and neurogenesis inhibition, together with increased blood–brain barrier permeability and gut dysbiosis, have been linked to ND pathogenesis. Since NDs still lack curative treatment, recent research has sought therapeutic options in restoring gut microbiota and supplementing probiotic bacteria-derived metabolites with beneficial action to the host—so called postbiotics. The current review focuses on literature explaining cellular mechanisms involved in ND pathogenesis and research addressing the impact that postbiotics as a whole mixture and particular metabolites, such as short-chain fatty acids (SCFAs), lactate, polyamines, polyphenols, tryptophan metabolites, exopolysaccharides and bacterial extracellular vesicles, have on the ageing-associated processes underlying ND occurrence. The review also discusses the issue of implementing postbiotics into ND prophylaxis and therapy, depicting them as compounds addressing senescence-triggered dysfunctions that are worth translating from bench to pharmaceutical market in response to “silver consumers” demands. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota and Neuropsychiatric Diseases)
Show Figures

Figure 1

14 pages, 649 KiB  
Review
Blood Microbiota and Its Products: Mechanisms of Interference with Host Cells and Clinical Outcomes
by Luigi Santacroce, Ioannis Alexandros Charitos, Marica Colella, Raffaele Palmirotta and Emilio Jirillo
Hematol. Rep. 2024, 16(3), 440-453; https://doi.org/10.3390/hematolrep16030043 - 6 Jul 2024
Cited by 2 | Viewed by 2424
Abstract
In healthy conditions, blood was considered a sterile environment until the development of new analytical approaches that allowed for the detection of circulating bacterial ribosomal DNA. Currently, debate exists on the origin of the blood microbiota. According to advanced research using dark field [...] Read more.
In healthy conditions, blood was considered a sterile environment until the development of new analytical approaches that allowed for the detection of circulating bacterial ribosomal DNA. Currently, debate exists on the origin of the blood microbiota. According to advanced research using dark field microscopy, fluorescent in situ hybridization, flow cytometry, and electron microscopy, so-called microbiota have been detected in the blood. Conversely, others have reported no evidence of a common blood microbiota. Then, it was hypothesized that blood microbiota may derive from distant sites, e.g., the gut or external contamination of blood samples. Alteration of the blood microbiota’s equilibrium may lead to dysbiosis and, in certain cases, disease. Cardiovascular, respiratory, hepatic, kidney, neoplastic, and immune diseases have been associated with the presence of Gram-positive and Gram-negative bacteria and/or their products in the blood. For instance, lipopolysaccharides (LPSs) and endotoxins may contribute to tissue damage, fueling chronic inflammation. Blood bacteria can interact with immune cells, especially with monocytes that engulf microorganisms and T lymphocytes via spontaneous binding to their membranes. Moreover, LPSs, extracellular vesicles, and outer membrane vesicles interact with red blood cells and immune cells, reaching distant organs. This review aims to describe the composition of blood microbiota in healthy individuals and those with disease conditions. Furthermore, special emphasis is placed on the interaction of blood microbiota with host cells to better understand disease mechanisms. Full article
Show Figures

Figure 1

Back to TopTop