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
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (137)

Search Parameters:
Keywords = Bifidobacterium Clostridium

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 3012 KiB  
Article
Investigating Multi-Omic Signatures of Ethnicity and Dysglycaemia in Asian Chinese and European Caucasian Adults: Cross-Sectional Analysis of the TOFI_Asia Study at 4-Year Follow-Up
by Saif Faraj, Aidan Joblin-Mills, Ivana R. Sequeira-Bisson, Kok Hong Leiu, Tommy Tung, Jessica A. Wallbank, Karl Fraser, Jennifer L. Miles-Chan, Sally D. Poppitt and Michael W. Taylor
Metabolites 2025, 15(8), 522; https://doi.org/10.3390/metabo15080522 - 1 Aug 2025
Viewed by 292
Abstract
Background: Type 2 diabetes (T2D) is a global health epidemic with rising prevalence within Asian populations, particularly amongst individuals with high visceral adiposity and ectopic organ fat, the so-called Thin-Outside, Fat-Inside phenotype. Metabolomic and microbiome shifts may herald T2D onset, presenting potential biomarkers [...] Read more.
Background: Type 2 diabetes (T2D) is a global health epidemic with rising prevalence within Asian populations, particularly amongst individuals with high visceral adiposity and ectopic organ fat, the so-called Thin-Outside, Fat-Inside phenotype. Metabolomic and microbiome shifts may herald T2D onset, presenting potential biomarkers and mechanistic insight into metabolic dysregulation. However, multi-omics datasets across ethnicities remain limited. Methods: We performed cross-sectional multi-omics analyses on 171 adults (99 Asian Chinese, 72 European Caucasian) from the New Zealand-based TOFI_Asia cohort at 4-years follow-up. Paired plasma and faecal samples were analysed using untargeted metabolomic profiling (polar/lipid fractions) and shotgun metagenomic sequencing, respectively. Sparse multi-block partial least squares regression and discriminant analysis (DIABLO) unveiled signatures associated with ethnicity, glycaemic status, and sex. Results: Ethnicity-based DIABLO modelling achieved a balanced error rate of 0.22, correctly classifying 76.54% of test samples. Polar metabolites had the highest discriminatory power (AUC = 0.96), with trigonelline enriched in European Caucasians and carnitine in Asian Chinese. Lipid profiles highlighted ethnicity-specific signatures: Asian Chinese showed enrichment of polyunsaturated triglycerides (TG.16:0_18:2_22:6, TG.18:1_18:2_22:6) and ether-linked phospholipids, while European Caucasians exhibited higher levels of saturated species (TG.16:0_16:0_14:1, TG.15:0_15:0_17:1). The bacteria Bifidobacterium pseudocatenulatum, Erysipelatoclostridium ramosum, and Enterocloster bolteae characterised Asian Chinese participants, while Oscillibacter sp. and Clostridium innocuum characterised European Caucasians. Cross-omic correlations highlighted negative correlations of Phocaeicola vulgatus with amino acids (r = −0.84 to −0.76), while E. ramosum and C. innocuum positively correlated with long-chain triglycerides (r = 0.55–0.62). Conclusions: Ethnicity drove robust multi-omic differentiation, revealing distinctive metabolic and microbial profiles potentially underlying the differential T2D risk between Asian Chinese and European Caucasians. Full article
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)
Show Figures

Figure 1

13 pages, 4616 KiB  
Article
Effect of Benzoic Acid on Nutrient Digestibility and Rectal Microbiota of Weaned Holstein Dairy Calves
by Haonan Dai, Dewei Du, Qi Huang, Jia Guo, Shujing Li, Wenli Yu, Zengyuan Zhao and Peng Sun
Animals 2025, 15(14), 2080; https://doi.org/10.3390/ani15142080 - 14 Jul 2025
Viewed by 393
Abstract
Our previous study has shown that supplementation of 0.50% benzoic acid (BA) increased growth performance, promoted rumen fermentation, and improved the composition and function of rumen microbiota. This research was designed to conduct a deeper exploration of the impacts of dietary supplementation with [...] Read more.
Our previous study has shown that supplementation of 0.50% benzoic acid (BA) increased growth performance, promoted rumen fermentation, and improved the composition and function of rumen microbiota. This research was designed to conduct a deeper exploration of the impacts of dietary supplementation with BA on the apparent digestibility of nutrients and the composition of rectal microbiota in weaned Holstein dairy calves. Sixteen Holstein heifer calves with similar body weights (91.2 ± 0.7 kg) were selected and randomly allocated into two groups, each comprising eight calves. Calves in the control group (CON group) were fed with a basal diet, while those in the benzoic acid group (BA group) were fed with the basal diet supplemented with 0.50% benzoic acid (on a dry matter basis). The experimental period started at 60 days of age and ended at 102 days of age, lasting for a total of 42 days. The calves were weaned at 60 days of age, with a transition period of 7 days. Feed samples were collected every two weeks, fecal samples were collected from 99 to 101 days of age, and blood samples were collected at 102 days of age. The results showed that supplementation with BA did not influence the digestibility of dry matter, crude protein, ether extract, neutral detergent fiber, acid detergent fiber, calcium, and phosphorus between the two groups. Compared with the CON group, BA supplementation tended to decrease the total cholesterol (TC) in the serum of the calves (p = 0.067). Supplementation with BA increased the relative abundances of the two beneficial bacteria, Bifidobacterium and Bifidobacterium pseudolongum (p < 0.05, LDA > 2), but decreased that of the harmful bacterium, Clostridium sensu stricto 1, in the rectum of dairy calves. The microbial functional prediction revealed that the fecal microbial metabolism involved in primary bile acid biosynthesis was higher in the calves from the BA group. In conclusion, the present study demonstrated that adding 0.50% BA to the diet did not influence the apparent nutrient digestibility, but improved rectal microbiota health, which finally promoted the growth performance in weaned Holstein dairy calves. Full article
(This article belongs to the Section Animal Nutrition)
Show Figures

Figure 1

17 pages, 847 KiB  
Review
Mechanistic Links Between Gut Dysbiosis, Insulin Resistance, and Autism Spectrum Disorder
by Patricia Guevara-Ramírez, Rafael Tamayo-Trujillo, Viviana A. Ruiz-Pozo, Santiago Cadena-Ullauri, Elius Paz-Cruz and Ana Karina Zambrano
Int. J. Mol. Sci. 2025, 26(13), 6537; https://doi.org/10.3390/ijms26136537 - 7 Jul 2025
Viewed by 949
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition frequently associated with gastrointestinal symptoms, gut dysbiosis, and metabolic dysfunctions such as insulin resistance (IR). Recent evidence suggests that the gut microbiota may influence both metabolic and neurological processes through the gut–brain–metabolic axis. This review [...] Read more.
Autism spectrum disorder (ASD) is a neurodevelopmental condition frequently associated with gastrointestinal symptoms, gut dysbiosis, and metabolic dysfunctions such as insulin resistance (IR). Recent evidence suggests that the gut microbiota may influence both metabolic and neurological processes through the gut–brain–metabolic axis. This review explores the molecular mechanisms linking dysbiosis, IR, and ASD, focusing on pathways such as TLR/NF-κB activation, PI3K/Akt/mTOR disruption, and the action of microbial metabolites, like short-chain fatty acids (SCFAs), lipopolysaccharide (LPS), and γ-aminobutyric acid (GABA). We discuss how dysbiosis may contribute to increased intestinal permeability, systemic inflammation, and neuroimmune activation, ultimately affecting brain development and behavior. Common microbial alterations in ASD and IR—including increased Clostridium, Desulfovibrio, and Alistipes, and reduced Bifidobacterium and butyrate-producing genera—suggest a shared pathophysiology. We also highlight potential therapeutic strategies, such as microbiota modulation, insulin-like growth factor 1 (IGF-1) treatment, and dietary interventions. Understanding these interconnected mechanisms may support the development of microbiota-targeted approaches for individuals with ASD metabolic comorbidities. Full article
(This article belongs to the Special Issue The Molecular and Cellular Aspects of Insulin Resistance)
Show Figures

Figure 1

21 pages, 1587 KiB  
Review
Microbiota—A Rescuing Modulator in Children Struggling with Functional Constipation
by Nicoleta Ana Tomșa, Lorena Elena Meliț, Teodora Popescu, Karina Najjar, Anca Meda Văsieșiu, Adrian Vlad Pop and Reka Borka-Balas
Microorganisms 2025, 13(7), 1504; https://doi.org/10.3390/microorganisms13071504 - 27 Jun 2025
Viewed by 580
Abstract
Constipation affects around 30% of children and in 95% of cases is functional (FC), a consequence of alterations in digestive tract peristalsis, modulated by the immune and nervous systems, bile acid metabolism, and the gut microbiota. The aim of this review was to [...] Read more.
Constipation affects around 30% of children and in 95% of cases is functional (FC), a consequence of alterations in digestive tract peristalsis, modulated by the immune and nervous systems, bile acid metabolism, and the gut microbiota. The aim of this review was to assess the role of gut microbiota and the use of probiotics in children with constipation. The current treatment involves education, toilet training, and oral laxatives, effective in only 50% of patients. In chronic FC, the composition of the microbiota is altered, with increased abundance of Bacteroidetes, Enterobacteriaceae, and Firmicutes and decreases in Prevotella, Bifidobacteria, Faecalibacterium prausnitzii, and Clostridium leptum. Probiotics replenish lacking beneficial resident bacteria, downregulate mucosal inflammation, or produce short-chain fatty acids (SCFAs). Probiotics like Bifidobacterium breve and Bifidobacterium longum increase the defecation frequency and decrease the episodes of both fecal incontinence and abdominal pain. Bifidobacterium animalis subsp. lactis XLTG11 improves the gut microbiota by upregulating SCFA genes and downregulating those related to methane metabolism. Lactobacilli produce organic acids that stimulate bowel peristalsis and augment fecal bolus moisture. The heterogeneity of the current studies involving pediatric subjects thus far hinders the use of probiotics as a standard in the management of children with constipation. Full article
(This article belongs to the Special Issue Microbiota and Gastrointestinal Diseases)
Show Figures

Figure 1

24 pages, 2904 KiB  
Article
Early Inoculation of a Multi-Species Probiotic in Piglets–Impacts on the Gut Microbiome and Immune Responses
by Lea Hübertz Birch Hansen, Charlotte Lauridsen, Bea Nielsen, Lisbeth Jørgensen, Anna Schönherz and Nuria Canibe
Microorganisms 2025, 13(6), 1292; https://doi.org/10.3390/microorganisms13061292 - 31 May 2025
Viewed by 775
Abstract
Intestinal diseases in nursery pigs harm health and performance and drive antimicrobial resistance. This study evaluated whether early probiotic inoculation helps piglets to cope with weaning-related gut challenges. The probiotic, containing Lacticaseibacillus rhamnosus, Enterococcus lactis, Bifidobacterium longum subsp. infantis, and [...] Read more.
Intestinal diseases in nursery pigs harm health and performance and drive antimicrobial resistance. This study evaluated whether early probiotic inoculation helps piglets to cope with weaning-related gut challenges. The probiotic, containing Lacticaseibacillus rhamnosus, Enterococcus lactis, Bifidobacterium longum subsp. infantis, and Bifidobacterium breve, was given orally to newborn piglets daily until day 4 and then every other day until weaning at day 28 (at 4 × 109 CFU/dose). The control piglets received a placebo. The results showed that the probiotic pigs had reduced fecal alpha-diversity on day 7 but greater Shannon diversity on day 28 (feces) and day 23 (intestinal contents) compared to those of the control pigs. Beta-diversity analysis showed microbial differences between the groups on day 35. Most zOTUs (zero-radius operational taxonomic units) found to significantly differentiate the two treatment groups were found pre weaning. Bifidobacterium breve, Ligilactobacillus salivarius, as well as Clostridium ramosum were significantly more abundant in the feces of the probiotic pigs more than once. The probiotic pigs had higher expression levels of mucin 2 (MUC2); solute carrier family 5, member 8 (SLC5A8); and interleukin 8 (IL-8) post weaning. In the early post-weaning period, the probiotic pigs had less diarrhea as well as lower cadaverine levels in digesta than the control pigs. In conclusion, early probiotic inoculation may induce lasting immunomodulation via microbial antigen changes, enhancing resilience during challenges, like weaning. Notably, the effects persisted beyond weaning and probiotic cessation. Full article
(This article belongs to the Section Veterinary Microbiology)
Show Figures

Figure 1

25 pages, 5953 KiB  
Article
Evaluation of the Functional Properties and Edible Safety of Concocted Xanthii Fructus Protein
by Yuchen Dong, Zihao Wan, Fuguo Han, Xuemei Fan, Yanli Hao, Fang Wei and Qingfei Liu
Foods 2025, 14(11), 1913; https://doi.org/10.3390/foods14111913 - 28 May 2025
Viewed by 540
Abstract
Xanthii Fructus (XF) not only has medicinal function in traditional Chinese medicine (TCM) but also contains rich oil and protein. The aim of this research was to develop the edible value of its protein based on the investigation on the extraction, basic characteristics [...] Read more.
Xanthii Fructus (XF) not only has medicinal function in traditional Chinese medicine (TCM) but also contains rich oil and protein. The aim of this research was to develop the edible value of its protein based on the investigation on the extraction, basic characteristics and functions, safety, gut microbiota, and metabolomics, especially the effect of the concocting process. The proteins from raw and concocted XF were prepared using two methods: alkaline solubilization followed by acid precipitation and ammonium sulfate salting-out, respectively. The secondary structure and physicochemical properties of the proteins were characterized through spectroscopic analysis and property determination. The effects of alkaline and the concocting process on the proteins were systematically compared. The results indicated that the salting-out method could retain the protein activity better. Both alkali treatment and the concocting process altered the folding state of proteins. The toxicological results in mice indicated that a high dose (0.35 g/kg) of raw Xanthii Fructus protein (XFP) might cause damage to the liver and small intestine, and the concocting process could significantly alleviate the damage. The 16S rRNA sequencing technology was used to untangle their impact on gut microbiota in mice and the result showed that raw protein had a certain regulatory effect on Bifidobacterium, Rhodococcus, Lactococcus, and Clostridium, while the concocted protein had a smaller impact, mainly affecting Bacteroides and Bifidobacterium. The untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS) showed that the proteins of raw XF affected the metabolic level through cysteine and methionine metabolism, purine metabolism, amino sugar and nucleotide sugar metabolism pathways, and the concocted protein mainly involved histidine metabolism and purine metabolism pathways. Overall, XFP had potential development prospects, but the anti-nutritional factors might have some toxicity. The concocting process could significantly improve its safety, and the concocted proteins were worth developing as a food source. In the future, the processing conditions should be further optimized and more systematic investigation should be performed to ensure the safety of XF as a food source. Full article
Show Figures

Figure 1

11 pages, 761 KiB  
Perspective
Tea Catechins: Potential Plant-Derived Feed Additives for Improving Chicken Intestinal Health and Productivity
by Bing Tian, Wenjing Zhuang, Yanle Fan, Yun Hu, Xiaoyan Cui, Tingting Li, Liyang Zhang, Xugang Luo and Shengchen Wang
Animals 2025, 15(11), 1553; https://doi.org/10.3390/ani15111553 - 26 May 2025
Viewed by 495
Abstract
In response to the global antimicrobial resistance and residue issues, most countries and regions have implemented comprehensive bans on the use of growth-promoting antibiotics in animal feed formulations. Catechins are a group of polyphenolic compounds that are naturally present in many fruits and [...] Read more.
In response to the global antimicrobial resistance and residue issues, most countries and regions have implemented comprehensive bans on the use of growth-promoting antibiotics in animal feed formulations. Catechins are a group of polyphenolic compounds that are naturally present in many fruits and plant-derived foods, emerging as potential antibiotic-alternative additives for promoting gut health and production performance in poultry farming. The compounds demonstrate dual biological functions: mitigating oxidative tissue damage and actively remodeling gut microbiota composition by suppressing enteric pathogens (e.g., Clostridium) and enriching beneficial bacteria (e.g., Bifidobacterium). Such multifaceted regulation enhances intestinal barrier integrity, thereby improving nutrient absorption and translating into measurable production benefits, including elevated growth rates and feed conversion efficiency. The current research suggests that the added levels of catechins in chicken diets ranges from 40 to 600 mg/kg. However, the research and application of catechins in chicken production are still at the initial stage, which requires more attention worldwide and further mechanism exploration. This mini-review synthesizes recent research findings on the effects of catechins on chicken health, aiming to promote the formulation of feed additive strategies using plant-derived compounds such as catechins as substitutes for antibiotics, to maintain the health of poultry and other animals while preserving production performance. Full article
Show Figures

Figure 1

20 pages, 3056 KiB  
Review
Artificial Sweeteners: A Double-Edged Sword for Gut Microbiome
by Helal F. Hetta, Nizar Sirag, Hassabelrasoul Elfadil, Ayman Salama, Sara F. Aljadrawi, Amani J. Alfaifi, Asma N. Alwabisi, Bothinah M. AbuAlhasan, Layan S. Alanazi, Yara A. Aljohani, Yasmin N. Ramadan, Noura H. Abd Ellah and Abdelazeem M. Algammal
Diseases 2025, 13(4), 115; https://doi.org/10.3390/diseases13040115 - 15 Apr 2025
Cited by 2 | Viewed by 8588
Abstract
Background and Aim: The human gut microbiome plays a crucial role in maintaining health. Artificial sweeteners, also known as non-nutritive sweeteners (NNS), have garnered attention for their potential to disrupt the balance of the gut microbiome. This review explores the complex relationship [...] Read more.
Background and Aim: The human gut microbiome plays a crucial role in maintaining health. Artificial sweeteners, also known as non-nutritive sweeteners (NNS), have garnered attention for their potential to disrupt the balance of the gut microbiome. This review explores the complex relationship between NNS and the gut microbiome, highlighting their potential benefits and risks. By synthesizing current evidence, we aim to provide a balanced perspective on the role of AS in dietary practices and health outcomes, emphasizing the need for targeted research to guide their safe and effective use. Methods: A comprehensive literature review was conducted through searches in PubMed and Google Scholar, focusing on the effects of artificial sweeteners on gut microbiota. The search utilized key terms including “Gut Microbiome”, “gut microbiota”, “Eubiosis”, “Dysbiosis”, “Artificial Sweeteners”, and “Nonnutritive Sweeteners”. Results: NNS may alter the gut microbiome, but findings remain inconsistent. Animal studies often report a decrease in beneficial bacteria like Bifidobacterium and Lactobacillus, and an increase in harmful strains such as Clostridium difficile and E. coli, potentially leading to inflammation and gut imbalance. Disruptions in short-chain fatty acid (SCFA) production and gut hormone signaling have also been observed. However, human studies generally show milder or no significant changes, highlighting the limitations in translating animal model findings directly to humans. Differences in study design, dosage, exposure time, and sweetener type likely contribute to these varied outcomes. Conclusions: While NNS offer certain benefits, including reduced caloric intake and improved blood sugar regulation, their impact on gut microbiome health raises important concerns. The observed reduction in beneficial bacteria and the rise in pathogenic strains underscore the need for caution in NNS consumption. Furthermore, the disruption of SCFA production and metabolic pathways illustrates the intricate relationship between diet and gut health. Full article
(This article belongs to the Special Issue Microbiota in Human Disease)
Show Figures

Figure 1

13 pages, 2416 KiB  
Review
Insights into the Interaction Between Clostridioides difficile and the Gut Microbiome
by Dimitra Mougiou, Georgia Gioula, Lemonia Skoura, Cleo Anastassopoulou and Melania Kachrimanidou
J. Pers. Med. 2025, 15(3), 94; https://doi.org/10.3390/jpm15030094 - 28 Feb 2025
Cited by 3 | Viewed by 1733
Abstract
Clostridioides difficile (C. difficile) is a significant healthcare-associated pathogen that is predominantly caused by antibiotic-induced microbiota disturbance. Antibiotics decrease microbial diversity, resulting in C. difficile colonization and infection. Clostridium difficile infection (CDI) manifests through toxins A and B, causing diarrhea and [...] Read more.
Clostridioides difficile (C. difficile) is a significant healthcare-associated pathogen that is predominantly caused by antibiotic-induced microbiota disturbance. Antibiotics decrease microbial diversity, resulting in C. difficile colonization and infection. Clostridium difficile infection (CDI) manifests through toxins A and B, causing diarrhea and colitis. Antibiotic usage, old age, and hospitalization are significant risk factors. A healthy gut microbiota, which is dominated by Firmicutes and Bacteroidetes, provides colonization resistance to C. difficile due to competition for nutrients, creating inhibitory substances and stimulating the immune response. Antibiotic-induced dysbiosis decreases resistance, allowing C. difficile spores to transform into vegetative forms. Patients with CDI have decreased gut microbiota diversity, with a decrease in beneficial bacteria, including Bacteroidetes, Prevotella, and Bifidobacterium, and a rise in harmful bacteria like Clostridioides and Lactobacillus. This disparity worsens the infection’s symptoms and complicates therapy. Fecal Microbiota Transplantation (FMT) has emerged as a potential therapy for recurrent CDI by restoring gut microbiota diversity and function. Comprehending the connection between gut microbiota and CDI pathogenesis is critical for establishing effective preventive and treatment plans. Maintaining a healthy gut microbiota through careful antibiotic use and therapeutic options such as FMT can help in the management and prevention of CDI. Full article
(This article belongs to the Special Issue Personalized Medicine in Infectious Diseases)
Show Figures

Figure 1

21 pages, 938 KiB  
Systematic Review
Gut Dysbiosis and Adult Atopic Dermatitis: A Systematic Review
by Kevin Díez-Madueño, Pablo de la Cueva Dobao, Isabel Torres-Rojas, Marta Fernández-Gosende, Claudio Hidalgo-Cantabrana and Pablo Coto-Segura
J. Clin. Med. 2025, 14(1), 19; https://doi.org/10.3390/jcm14010019 - 24 Dec 2024
Cited by 2 | Viewed by 3442
Abstract
Background/Objectives: Research on the relationship between gut microbiota (GM) and atopic dermatitis (AD) has seen a growing interest in recent years. The aim of this systematic review was to determine whether differences exist between the GM of adults with AD and that of [...] Read more.
Background/Objectives: Research on the relationship between gut microbiota (GM) and atopic dermatitis (AD) has seen a growing interest in recent years. The aim of this systematic review was to determine whether differences exist between the GM of adults with AD and that of healthy adults (gut dysbiosis). Methods: We conducted a systematic review based on the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). The search was performed using PubMed, EMBASE, and Web of Science. Observational and interventional studies were analyzed. Results: Although the studies showed heterogeneous results, some distinguishing characteristics were found in the intestinal microbial composition of adults with dermatitis. Even though no significant differences in diversity were found between healthy and affected adults, certain microorganisms, such as Bacteroidales, Enterobacteriaceae, and Clostridium (perfringens), were more characteristic of the fecal microbiota in adults with AD. Healthy individuals exhibited lower abundances of aerobic bacteria and higher abundances of short-chain fatty acid-producing species and polyamines. Clinical trials showed that the consumption of probiotics (Bifidobacterium and/or Lactobacillus), fecal microbiota transplants, and balneotherapy modified the fecal microbiota composition of participants and were associated with significant improvements in disease management. Conclusions: In anticipation of forthcoming clinical trials, it is essential to conduct meta-analyses that comprehensively evaluate the effectiveness and safety of interventions designed to modify intestinal flora in the context of AD. Preliminary evidence suggests that certain interventions may enhance adult AD management. Full article
(This article belongs to the Special Issue New Advances in Alopecia Areata and Atopic Dermatitis)
Show Figures

Figure 1

18 pages, 2274 KiB  
Review
Application of Invasive Bacteria for the Delivery of Anti-Cancer Therapeutics
by Rasaq Akinsola and Kumaran Narayanan
Therapeutics 2024, 1(2), 124-141; https://doi.org/10.3390/therapeutics1020011 - 20 Dec 2024
Cited by 1 | Viewed by 2150
Abstract
Bacterial vectors for biomolecule delivery to targeted organelles, facilitating temporary or continuous protein production, have emerged as a promising approach for treating acquired and inherited diseases. This method offers a selective cancer eradication and targeting strategy with minimal side effects. Bacterial vectors provide [...] Read more.
Bacterial vectors for biomolecule delivery to targeted organelles, facilitating temporary or continuous protein production, have emerged as a promising approach for treating acquired and inherited diseases. This method offers a selective cancer eradication and targeting strategy with minimal side effects. Bacterial vectors provide an alternative to viral gene delivery, given their capacity to deliver large genetic materials while inducing minimal immunogenicity and cytotoxicity. Bacteria such as Bifidobacterium, Salmonella, Clostridium, and Streptococcus have demonstrated potential for tumor-targeted biomolecule delivery or serve as oncolytic bacteria. These vectors have also been used to transfer and amplify genes encoding biomolecules such as pro-drug-converting enzymes, toxins, angiogenesis inhibitors, and cytokines. The microenvironment of necrotic tumors offers a unique opportunity for targeted therapy with the non-pathogenic anaerobic bacterium. For example, Clostridium sporogenes can germinate selectively in the necrotic regions upon injection as endospores, which helps to enhance the specificity of Clostridium sporogenes, resulting in tumor-specific colonization. Also, E. coli and Salmonella sp. can be capacitated with a hypoxic sensing promotor gene for specificity delivery into the core region of solid tumors. The uniqueness of the tumor microenvironment, including hypoxia, immunosuppression, metabolite deficiency or enrichment, and necrosis, selectively enables bacteria in the tumor. Combining traditional cancer therapy with bacterial therapy will significantly complement and cover the limitations of other treatments. This review provides an overview of the use of the bacteria vector in cancer therapy, discussing strategies to maximize delivery efficiency and address potential challenges. In this review, we discuss the potential of bacteria vectors as anti-cancer therapeutics while focusing on therapeutic delivery strategies. We highlight the complementary use of bacteria therapy with other cancer therapies and the mechanism of bacteria cancer immunotherapy with limitations and perspectives for future use. Full article
Show Figures

Figure 1

31 pages, 432 KiB  
Review
Promising Probiotic Candidates for Sustainable Aquaculture: An Updated Review
by Seyed Hossein Hoseinifar, Mehwish Faheem, Iram Liaqat, Hien Van Doan, Koushik Ghosh and Einar Ringø
Animals 2024, 14(24), 3644; https://doi.org/10.3390/ani14243644 - 17 Dec 2024
Cited by 5 | Viewed by 3658
Abstract
With the intensification of aquaculture to meet the rising demands of fish and shellfish, disease outbreaks during the larval and adult stages are a major challenge faced by aqua culturists. As the prophylactic use of vaccines and antibiotics has several limitations, research is [...] Read more.
With the intensification of aquaculture to meet the rising demands of fish and shellfish, disease outbreaks during the larval and adult stages are a major challenge faced by aqua culturists. As the prophylactic use of vaccines and antibiotics has several limitations, research is now focused on sustainable alternatives to vaccines and antibiotics, e.g., medicinal plants, probiotics, postbiotics, prebiotics, and synbiotics, as promising candidates to strengthen the immune response of fish and shellfish and to control disease outbreaks. With respect to probiotics, numerous studies are available revealing their health-promoting and beneficial impacts in aquaculture. However, most studies focus on Bacillus and Lactobacillus species. Keeping in view the positive effects of probiotic lactic acid bacteria in aquaculture, researchers are now looking for other probiotic bacteria that can be used in aquaculture. Recently, many non-lactic acid bacteria (non-LAB), which are mainly host-associated, have been reported to reveal beneficial effects in fish and shellfish aquaculture. The main non-LAB probiotic genera are Bifidobacterium, Clostridium, Microbacterium, Micrococcus, Paenibacillus, Acinetobacter, Alcaligenes, Enterobacter, Phaeobacter Pseudoalteromonas, Pseudomonas, Pseudomonas, and Vibrio. Despite the promising effects of non-LAB probiotics, comparably, there is limited available information in this context. This review focuses only on probiotic strains that are non-LAB, mostly isolated from the host digestive tract or rearing water, and discusses their beneficial effects in fish and shellfish aquaculture. This review will provide detailed information on the use of various non-LAB bacteria and provide a roadmap to future studies on new probiotics for sustainable aquaculture. Full article
(This article belongs to the Special Issue Gut Microbiota in Aquatic Animals)
13 pages, 1266 KiB  
Article
The Impact of Complementary Feeding on Fecal Microbiota in Exclusively Breast-Fed Infants with Cystic Fibrosis (A Descriptive Study)
by Andrea Asensio-Grau, María Garriga, Saioa Vicente, Ana Andrés, Carmen Ribes-Koninckx and Joaquim Calvo-Lerma
Nutrients 2024, 16(23), 4071; https://doi.org/10.3390/nu16234071 - 27 Nov 2024
Viewed by 1094
Abstract
Background/Objectives: Early life gut microbiota plays a pivotal role in shaping immunity, metabolism, and overall health outcomes. This is relevant in healthy infants but may be even more crucial in infants with chronic devastating diseases, such as cystic fibrosis (CF). While the introduction [...] Read more.
Background/Objectives: Early life gut microbiota plays a pivotal role in shaping immunity, metabolism, and overall health outcomes. This is relevant in healthy infants but may be even more crucial in infants with chronic devastating diseases, such as cystic fibrosis (CF). While the introduction of solid foods in healthy infants modifies the composition of colonic microbiota, less knowledge is available on those with CF. The aim of this descriptive observational study was to assess the composition of fecal microbiota in six exclusively breast-fed infants with CF, and then explore the changes induced upon the introduction of different foods. Methods: two types of fecal samples were collected from each subject: one during the exclusive-breastfeeding period, and the other after incorporating each new food in the ad libitum diet. The microbiota composition was analyzed by 16S rRNA amplicon sequencing. Results: Wide heterogenicity in the composition at the phylum level (variable proportions of Actinobacteriota, Proteobacteria, and Firmicutes, and the absence of Bacteroidota in all subjects) was found, and different enterotypes were characterized in each subject by the main presence of one genus: Bifidobacterium in Subject 1 (relative abundance of 54.4%), Klebsiella in Subject 3 (49.1%), Veillonella in Subjects 4 and 5 (32.7% and 36.9%, respectively), and Clostridium in Subject 6 (48.9%). The transition to complementary feeding induced variable changes in microbiota composition, suggesting a subject-specific response and highlighting the importance of inter-individual variation. Conclusions: Further studies are required to identify which foods contribute to shaping colonic microbiota in the most favorable way for patients with CF using a personalized approach. Full article
(This article belongs to the Special Issue Nutrition and Cystic Fibrosis in Children)
Show Figures

Figure 1

11 pages, 2943 KiB  
Article
Comparison of the Gut Microbiota of Patients Who Improve with Antibiotic Combination Therapy for Ulcerative Colitis and Those Who Do Not: Investigation by Fecal Metagenomic Analyses
by Toshifumi Ohkusa, Kimitoshi Kato, Tsuyoshi Sekizuka, Toshiro Sugiyama, Nobuhiro Sato and Makoto Kuroda
Nutrients 2024, 16(20), 3500; https://doi.org/10.3390/nu16203500 - 16 Oct 2024
Cited by 2 | Viewed by 2030
Abstract
Background/Objectives: The cause of ulcerative colitis (UC) may be related to commensal bacteria in genetically susceptible patients. We previously demonstrated that triple antibiotic combination therapy induces remission in patients with active UC in randomized controlled trials (RCTs). Now, we investigate changes in [...] Read more.
Background/Objectives: The cause of ulcerative colitis (UC) may be related to commensal bacteria in genetically susceptible patients. We previously demonstrated that triple antibiotic combination therapy induces remission in patients with active UC in randomized controlled trials (RCTs). Now, we investigate changes in the gut microbiota of patients who responded to the antibiotic combination therapy. Methods: Thirty-one patients with UC given ATM/AFM (amoxicillin, metronidazole, and tetracycline or fosfomycin) therapy for two weeks were enrolled in this study. The clinical conditions of these UC patients were evaluated by the partial Mayo score. The gut microbiota was compared via the metagenomic shot gun analysis of fecal samples. Results: Of the 31 patients, 16 and 8 experienced complete and partial remission, respectively, over three months in response to ATM/AFM therapy, whereas ATM/AFM showed no efficacy in 7 patients. The dysbiosis before treatment in the active stage could be associated with increased populations of Bacteroides, Parabacteroides, Rickenella, Clostridium, Flavonifractor, Pelagibacter, Bordetella, Massilia, and Piscrickettsia species. Metagenomic analysis revealed dramatic changes in the gut microbiota at an early stage, that is, just two weeks after starting ATM/AFM therapy. After treatment in the responder group, the populations of bifidobacterium and lactobacilli species were significantly increased, while the population of bacteroides decreased. Conclusions: These results suggest that metagenomic analysis demonstrated a marked change in the gut microbiota after antibiotic combination treatment. In the triple antibiotic combination therapy, remission was associated with an increase in bifidobacterium and lactobacilli species. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Figure 1

16 pages, 6521 KiB  
Article
The Composition and Function of Intestinal Microbiota Were Altered in Farmed Bullfrog Tadpoles (Aquarana catesbeiana) during Metamorphosis
by Xiaoting Zheng, Qiuyu Chen, Xueying Liang, Vikash Kumar, Alfredo Loor, Hongbiao Dong, Chang Liu, Jinlong Yang and Jiasong Zhang
Microorganisms 2024, 12(10), 2020; https://doi.org/10.3390/microorganisms12102020 - 5 Oct 2024
Cited by 1 | Viewed by 1465
Abstract
The bullfrog Aquarana catesbeiana is one of the main farmed frog species in China, with a low overall survival of farmed bullfrogs from hatching to harvest since bullfrog tadpoles are fragile during the metamorphosis period. The intestinal bacterial community can play crucial roles [...] Read more.
The bullfrog Aquarana catesbeiana is one of the main farmed frog species in China, with a low overall survival of farmed bullfrogs from hatching to harvest since bullfrog tadpoles are fragile during the metamorphosis period. The intestinal bacterial community can play crucial roles in animal development; however, little is known about the alteration of the gut microbial community of A. catesbeiana during metamorphosis. The present study used 16S rRNA amplicon sequencing to investigate the intestinal bacterial community in A. catesbeiana at four distinct developmental stages. Moreover, we determined the bullfrog’s body morphological parameters and the intestine histology at different developmental stages. The results showed a reduction in the total length and snout–vent length of A. catesbeiana during metamorphosis. The intestinal microbial composition of A. catesbeiana exhibited variation throughout the process of metamorphosis. The terrestrial stage showed shifts in the bacterial composition compared to the aquatic stages, including a reduction in Bacteroidetes and an increase in Firmicutes. Furthermore, the presence of Prevotella, Bifidobacterium, Leucobacter, Corynebacterium, Bulleidia, Dorea, Robinsoniella, and Clostridium in A. catesbeiana metamorphosis appears to be mainly related to the host’s epithelial cells’ height and total body mass. The results indicated that the intestinal microbial composition changed with the bullfrog–tadpole metamorphosis. The genera of Prevotella, Bifidobacterium, Leucobacter, Corynebacterium, Bulleidia, Dorea, Robinsoniella, and Clostridium might be potential probiotics. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Back to TopTop