1
|
Harris RG, Neale EP, Batterham M. Efficacy of Probiotics Compared With Pharmacological Treatments for Maintenance Therapy for Functional Constipation in Children: A Systematic Review and Network Meta-analysis. Nutr Rev 2025; 83:1006-1034. [PMID: 39348282 PMCID: PMC12066947 DOI: 10.1093/nutrit/nuae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024] Open
Abstract
CONTEXT There has been an increase in randomized controlled trials (RCTs) comparing probiotics with various maintenance therapies, such as polyethylene glycol, lactulose, and mineral oil, to treat functional constipation in children. OBJECTIVE The aim was to compare probiotics with all other oral maintenance therapies for functional constipation in children and rank all treatments in terms of effectiveness in a network meta-analysis. METHODS RCTs were identified through systematically searching the MEDLINE, Scopus, EMBASE, and Cochrane Library databases, trial registries, and forward and backward citation searching. Within-study risk of bias was assessed using the Cochrane Risk of Bias 2 tool, and confidence in the estimates was assessed using the CINeMA (Confidence in Network Meta-Analysis) framework. Random-effects network meta-analyses were conducted. RESULTS Data were pooled from 41 and 29 RCTs for network meta-analysis of defecation frequency and treatment success, respectively. Probiotics did not significantly increase the number of bowel movements per week when compared with any conventional treatment or placebo. A combination of mineral oil and probiotics was the most effective treatment for increasing defecation frequency (mean difference: 3.13; 95% confidence interval [CI]: 0.64, 5.63). The most effective treatments for increasing the risk of treatment success as compared with placebo were mineral oil (relative risk [RR]: 2.41; 95% CI: 1.53, 3.81) and a combined treatment of polyethylene glycol and lactulose (RR: 2.45; 95% CI: 1.21, 4.97). Confidence in the estimates ranged from very low to moderate. CONCLUSION Currently, there is no evidence to suggest that probiotics should be used as a standalone treatment for functional constipation in children. More high-quality studies are needed to evaluate different strains of probiotics and their potential benefit as an additional treatment component to conventional treatments. Mineral oil and polyethylene glycol were the most effective treatments to increase defecation frequency and treatment success rates and should remain the first line of treatment for children with functional constipation. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022360977 (https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=360977).
Collapse
Affiliation(s)
- Rebecca G Harris
- School of Medical Indigenous and Health Science, Faculty of Science, Medicine, and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia
| | - Elizabeth P Neale
- School of Medical Indigenous and Health Science, Faculty of Science, Medicine, and Health, University of Wollongong, Wollongong, New South Wales 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia
| | - Marijka Batterham
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia
- Statistical Consulting Centre, School of Mathematics and Applied Statistics, Faculty of Engineering and Information Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia
| |
Collapse
|
2
|
Plank BCA, Guergoletto KB, Rocha TS. Improved Bacterial Survival and Antioxidant Activity After In Vitro Digestion of Fermented Dairy Beverages by Lacticaseibacillus casei LC-01 and Lactiplantibacillus plantarum BG-112 Containing Yacon. Probiotics Antimicrob Proteins 2025; 17:1584-1595. [PMID: 38326639 DOI: 10.1007/s12602-024-10220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
The fermentation of milk containing 0%, 3%, 6%, and 9% (w/v) yacon root flour (YRF) by Lacticaseibacillus casei LC-1 and Lactiplantibacillus plantarum BG-112 was evaluated for bacterial survival and antioxidant activity (AA) before and after simulated gastrointestinal digestion. After 28 days of refrigerated storage, samples of the beverages were analyzed for cell viability, AA (using ferric ion reducing antioxidant power (FRAP) and (ABTS), and molecular mass profile of proteins (using electrophoresis). The presence of 9% YRF increased bacterial survival during 28 days of storage and passage through the gastrointestinal tract for both L. casei and L. plantarum, which showed a greater capacity to reduce ferric ions compared to 0% YRF, and the ability to capture free radicals increased from below 5 mM to over 15 mM TE after digestion. Milk proteins are hydrolyzed during digestion, and the generation of bioactive peptides with AA may explain the increase in AA levels. Since peptides are generated from milk proteins, YRF did not influence AA after digestion. These results showed that fermentation of milk by L. casei and L. plantarum with YRF increased the chances of these bacteria reaching the colon in adequate quantities. After simulated digestion, the beverages showed improved AA due to milk protein hydrolysis.
Collapse
Affiliation(s)
- Bruna C A Plank
- Department of Food Science and Technology, State University of Londrina, Londrina, PR, Brazil
| | - Karla B Guergoletto
- Department of Food Science and Technology, State University of Londrina, Londrina, PR, Brazil
| | - Thais S Rocha
- Department of Food Science and Technology, State University of Londrina, Londrina, PR, Brazil.
| |
Collapse
|
3
|
Gupta MK, Srivastava R. Gut Microbiome Interventions: From Dysbiosis to Next-Generation Probiotics (NGPs) for Disease Management. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10582-7. [PMID: 40434505 DOI: 10.1007/s12602-025-10582-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
The gut microbiome, sometimes referred to as the "second brain," the "lost organ," the "identification card of the individual," and the "fingerprint of the host," possesses diverse traits and functions that influence health. The impact of gut commensal bacteria on health, as opposed to environmental pathogenic factors, has generated increasing interest in recent years, culminating in a substantial body of study. Research indicates that dysbiosis of the intestinal microbiota is commonly observed in chronic inflammatory diseases, including colitis, obesity/metabolic syndrome, diabetes mellitus, liver infections, allergic conditions, cardiovascular diseases, COVID-19, cancers, and neurodegenerative disorders. The International Scientific Association for Probiotics and Prebiotics has recently refined the theory of complementary and synergistic synbiotics. In recent years, the field of microbiome research has been significantly advanced by technological developments such as massive culturomics, gnotobiotics, metabolomics, parallel DNA sequencing, and RNA sequencing. This review article examined the potential next generation probiotics (NGPs) and explored some of them, Faecalibacterium prausnitzii, Bacteroides thetaiotaomicron, Akkermansia muciniphila, Parabacteroides goldsteinii, Bacteroides fragilis, Eubacterium hallii, Roseburia intestinalis, Christensenella minuta, Prevotella copri, and Oscillospira guilliermondii. In addition to these useful probiotic strains, psychobiotics, members of the families of Lactobacilli, Streptococci, Bifidobacteria, Escherichia, and Enterococci, have extended applicability in the use for neurodevelopmental and neurodegenerative disorders. The article also reviewed current trends and limitations in NGPs to enhance our comprehensive understanding of key concepts associated with the consumption of probiotics and proposed necessary initiatives for researchers to engage in collaborative translational research as future therapeutic solutions.
Collapse
Affiliation(s)
- Mandeep Kumar Gupta
- Moradabad Educational Trust Group of Institutions Faculty of Pharmacy, Moradabad, 244001, Uttar Pradesh, India.
| | - Rajnish Srivastava
- Chitkara University School of Pharmacy, Chitkara University, Baddi, 174103, Himachal Pradesh, India
| |
Collapse
|
4
|
Mugo CW, Church E, Horniblow RD, Mollan SP, Botfield H, Hill LJ, Sinclair AJ, Grech O. Unravelling the gut-brain connection: a systematic review of migraine and the gut microbiome. J Headache Pain 2025; 26:125. [PMID: 40399789 PMCID: PMC12096802 DOI: 10.1186/s10194-025-02039-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/16/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND There is substantial evidence linking migraines to gastrointestinal (GI) issues. Conditions such as irritable bowel syndrome and colitis often co-occur with migraines and GI symptoms are common among migraine patients. However, the evidence supporting the efficacy of gut microbiome-targeted therapies for managing migraines is limited. This systematic review aimed to describe the existing evidence of the gut microbiome in patients with migraine compared to healthy individuals. Additionally, it sought to examine how therapies targeting the gut microbiome including prebiotics, probiotics and synbiotics, might influence clinical outcomes. METHODS We performed searches on Embase, PubMed, and the Cochrane Library to identify studies in migraines and the gut microbiome, focusing on those which investigated the gut microbiome composition and gut microbiome-targeted therapies. Key data was extracted and analysed including study details, patient demographics, migraine type, comorbidities, and clinical outcomes. For gut microbiome composition studies, bacterial diversity and abundance was noted. For gut microbiome-targeted therapies studies, treatment types, dosages, and patient outcomes was recorded. RESULTS A significant difference between various genera of microbes was reported between migraine patients and controls in several studies. Bacteroidetes (also named Bacteroidota), proteobacteria, and firmicutes (also named Bacillota) phyla groups were found significantly abundant in migraine, while studies were conflicted in the abundance of Actinobacteria and Clostridia with regards to increased migraine risk in migraine patients. Patients with migraine had a gut microbiome with reduced species number and relative abundance, as well as a distinct bacterial composition compared to controls. Synbiotic and synbiotic/probiotic combination treatments have been shown in five randomised controlled trials and one open label pilot study to significantly decrease migraine severity, frequency, duration and painkiller consumption. CONCLUSIONS The significant alterations in microbial phyla observed in migraine patients suggest a potential microbial signature that may be associated with migraine risk or chronic progression. However, the mechanistic underpinnings of these associations remain unclear. This systemic review found that probiotic and synbiotic/probiotic combination therapies may be promising interventions for migraine management, offering significant reductions in migraine frequency and painkiller use. Future randomised controlled studies are needed to evaluate the optimal length of treatment and impact on patient related quality of life.
Collapse
Affiliation(s)
- Caroline W Mugo
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ella Church
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Richard D Horniblow
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Susan P Mollan
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Neuro-Ophthalmology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Hannah Botfield
- Inflammation and Ageing, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lisa J Hill
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK
| | - Alexandra J Sinclair
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK.
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK.
| | - Olivia Grech
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
5
|
Ullah H. Gut-vitamin D interplay: key to mitigating immunosenescence and promoting healthy ageing. Immun Ageing 2025; 22:20. [PMID: 40390005 PMCID: PMC12087203 DOI: 10.1186/s12979-025-00514-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/26/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Immunosenescence is the loss and change of immunological organs, as well as innate and adaptive immune dysfunction with ageing, which can lead to increased sensitivity to infections, age-related diseases, and cancer. Emerging evidence highlights the role of gut-vitamin D axis in the regulation of immune ageing, influencing chronic inflammation and systemic health. This review aims to explore the interplay between the gut microbiota and vitamin D in mitigating immunosenescence and preventing against chronic inflammation and age-related diseases. MAIN TEXT Gut microbiota dysbiosis and vitamin D insufficiency accelerate immunosenescence and risk of chronic diseases. Literature data reveal that vitamin D modulates gut microbiota diversity and composition, enhances immune resilience, and reduce systemic inflammation. Conversely, gut microbiota influences vitamin D metabolism to promote the synthesis of active vitamin D metabolites with implications for immune health. CONCLUSIONS These findings underscore the potential of targeting gut-vitamin D axis to modulate immune responses, delay the immune ageing, and mitigate age-related diseases. Further research is needed to integrate vitamin D supplementation and microbiome modulation into strategies aimed at promoting healthy ageing.
Collapse
Affiliation(s)
- Hammad Ullah
- School of Pharmacy, University of Management and Technology, Lahore, 54000, Pakistan.
| |
Collapse
|
6
|
Iqbal A, Bokhari SFH, Rehman MU, Faizan Sattar SM, Bakht D, Dost W, Basit A. Gut-brain connection in schizophrenia: A narrative review. World J Psychiatry 2025; 15:103751. [DOI: 10.5498/wjp.v15.i5.103751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/23/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Schizophrenia is a complex neuropsychiatric disorder characterized by cognitive, emotional, and behavioral impairments. The microbiota-gut-brain axis is crucial in its pathophysiology, mediating communication between the gut and brain through neural, immune, endocrine, and metabolic pathways. Dysbiosis, or an imbalance in gut microbiota, is linked to neuroinflammation, systemic inflammation, and neurotransmitter disruptions, all of which contribute to the symptoms of schizophrenia. Gut microbiota-derived metabolites, such as short-chain fatty acids, influence brain function, including immune responses and neurotransmitter synthesis. These findings suggest that microbial imbalances exacerbate schizophrenia, providing a novel perspective on the disorder’s underlying mechanisms. Emerging microbiota-targeted therapies—such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation—show promise as adjunctive treatments, aiming to restore microbial balance and improve clinical outcomes. While further research is needed, targeting the microbiota-gut-brain axis offers an innovative approach to schizophrenia management, with the potential to enhance patient outcomes and quality of life.
Collapse
Affiliation(s)
- Asma Iqbal
- Department of Medicine and Surgery, King Edward Medical University, Lahore 54000, Punjab, Pakistan
| | | | - Muneeb Ur Rehman
- Department of Medicine and Surgery, King Edward Medical University, Lahore 54000, Punjab, Pakistan
| | | | - Danyal Bakht
- Department of Medicine and Surgery, King Edward Medical University, Lahore 54000, Punjab, Pakistan
| | - Wahidullah Dost
- Department of Curative Medicine, Kabul University of Medical Sciences, Kabul 10001, Afghanistan
| | - Abdul Basit
- Department of Medicine and Surgery, King Edward Medical University, Lahore 54000, Punjab, Pakistan
| |
Collapse
|
7
|
Alowo D, Olum S, Mukisa IM, Ongeng D. Prebiotic potential of oligosaccharides extracted from improved Ugandan varieties of millet, sesame, soybean, and sorghum: enhancing probiotic growth and enteric pathogen inhibition. BMC Microbiol 2025; 25:307. [PMID: 40389861 PMCID: PMC12087199 DOI: 10.1186/s12866-025-04028-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Functional gastrointestinal disorders like diarrhea continue to affect children under five years in low-income countries. Incorporating health-enhancing bioactive compounds such as prebiotics in diet offers a promising solution. This study investigated prebiotic potential of oligosaccharides extracted from improved varieties of millet (Seremi 2, Naromil 2), sesame (Sesim 2, Sesim 3), soybean (Maksoy 3N, Maksoy 6N), and sorghum (Narosorg 2, Narosorg 4), commonly consumed in Uganda. These were compared to their respective indigenous variety. This study employed standardized methods for optical density measurement, culture preparation, and oligosaccharide extraction to evaluate prebiotic properties. We investigated whether plant-based oligosaccharides could enhance the effectiveness of probiotics, specifically Lactiplantibacillus plantarum (ATCC 14917) and Lacticaseibacillus rhamnosus (ATCC 7469), in antagonizing common enteric pathogens (Salmonella enterica subsp. enterica (ATCC 13076) and Shigella flexneri (ATCC 12022)). Approximately 4-8 log CFU/ml of each probiotic was incubated in 2% w/v oligosaccharide extracts at 37 °C to evaluate the influence of the extracts on their growth, short-chain fatty acid (SCFA) production and antagonistic activity. Maximum cell density, which exceeded the minimum recommended probiotic cell density (6 log CFU/ml), was achieved during 24-h incubation period. The probiotics exhibited optimal growth in extracts of Sesim 2, Maksoy 3N, Narosorg 2 and indigenous millet variety resulting in a 68-84% increase in cell densities. The concentration of SCFA concentration was significantly higher (p < 0.05) in soybean-based oligosaccharides. Both probiotics antagonized growth of Salmonella and Shigella by more than 40% when cultured on Sesim 2, Maksoy 3N, Narosorg 2 and indigenous millet variety, while maintaining the probiotic cell densities above the minimum recommended level. These varieties show great potential as functional ingredients for developing synbiotic-rich foods to promote gut and public health. However, to evaluate the oligosaccharides prebiotic efficacy, in vitro fermentation using fecal microbiota and in vivo studies are necessary to determine gut microbiota changes and interactions.
Collapse
Affiliation(s)
- Docus Alowo
- Department of Food Science and Postharvest Technology, Faculty of Agriculture and Environment, Gulu University, P.O. Box, 166, Gulu, Uganda.
- Department of Food Innovation and Nutrition, Faculty of Agriculture and Environmental Sciences, Mountains of the Moon University, P.O. Box, 837, Fort Portal, Uganda.
| | - Solomon Olum
- Department of Food Science and Postharvest Technology, Faculty of Agriculture and Environment, Gulu University, P.O. Box, 166, Gulu, Uganda
| | - Ivan Muzira Mukisa
- Department of Food Technology & Nutrition, School of Food Technology, Nutrition & Bioengineering, College of Agricultural and Environmental Sciences, Makerere University, P.O. Box 7062, Kampala, Uganda
| | - Duncan Ongeng
- Department of Food Science and Postharvest Technology, Faculty of Agriculture and Environment, Gulu University, P.O. Box, 166, Gulu, Uganda
| |
Collapse
|
8
|
Reznik E, Newberry C. Current treatment options for adult patients with short gut syndrome: Do prebiotics, probiotics, and synbiotics play a role? Nutr Clin Pract 2025. [PMID: 40344579 DOI: 10.1002/ncp.11319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Short bowel syndrome (SBS) is a malabsorptive disorder characterized by reduced small intestinal length that results in nutrient deficiencies. Most commonly in adults, it occurs in the setting of surgical bowel resection related to trauma, infarction, or inflammatory bowel disease. Management principles include optimizing nutrition and hydration status via parenteral nutrition and oral diet and utilizing antisecretory, antimotility, and enterohormone agents to enhance intestinal absorption, facilitate intestinal adaptation, and reduce stool output. Other therapeutics include antibiotics to treat small intestinal bacterial overgrowth (SIBO) and microbial dysbiosis. Considering limitations to antibiotic use, in addition to a greater understanding of the nuances of the microbiome in digestive health regulation, there is also burgeoning interest in the role of prebiotics, probiotics, and synbiotics in SBS management. This review highlights current management principles of SBS with a broader discussion of clinical indications and safety considerations for use of prebiotics, probiotics, and synbiotics in this population. Although current research is in its infancy, prebiotics, probiotics, and synbiotics may represent a viable future therapeutic option in the management of this complex disorder, with further studies needed to define definitive regimens and update guidelines.
Collapse
Affiliation(s)
- Elizabeth Reznik
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Carolyn Newberry
- Division of Gastroenterology, Weill Cornell Medical Center, New York, New York, USA
| |
Collapse
|
9
|
Fang L, Peng H, Tan Z, Deng N, Peng X. The Role of Gut Microbiota on Intestinal Fibrosis in Inflammatory Bowel Disease and Traditional Chinese Medicine Intervention. J Inflamm Res 2025; 18:5951-5967. [PMID: 40357383 PMCID: PMC12067688 DOI: 10.2147/jir.s504827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the intestine, frequently complicated by intestinal fibrosis. As fibrosis progresses, it can result in luminal stricture and compromised intestinal function, significantly diminishing patients' quality of life. Emerging evidence suggests that gut microbiota and their metabolites contribute to the pathogenesis of IBD-associated intestinal fibrosis by influencing inflammation and modulating immune responses. This review systematically explores the mechanistic link between gut microbiota and intestinal fibrosis in IBD and evaluates the therapeutic potential of traditional Chinese medicine (TCM) interventions. Relevant studies were retrieved from PubMed, Web of Science, Embase, Scopus, CNKI, Wanfang, and VIP databases. Findings indicate that TCM, including Chinese herbal prescriptions and bioactive constituents, can modulate gut microbiota composition and microbial metabolites, ultimately alleviating intestinal fibrosis through anti-inflammatory, immunemodulatory, and anti-fibrotic mechanisms. These insights highlight the potential of TCM as a promising strategy for targeting gut microbiota in the management of IBD-associated fibrosis.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Huiyi Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
10
|
Liu Y, Hu Y, Ma B, Wang Z, Wei B. Gut Microbiota and Exercise: Probiotics to Modify the Composition and Roles of the Gut Microbiota in the Context of 3P Medicine. MICROBIAL ECOLOGY 2025; 88:38. [PMID: 40319213 PMCID: PMC12049406 DOI: 10.1007/s00248-025-02529-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/06/2025] [Indexed: 05/07/2025]
Abstract
Prolonged and intense physical activity can trigger stress response mechanisms across various physiological systems-including the cardiovascular, respiratory, gastrointestinal, musculoskeletal, and neuroendocrine systems-disrupting energy metabolism, immune function, redox balance, and hormonal regulation. Critically, when not accompanied by adequate recovery, such exertion may impair rather than enhance athletic performance. In parallel, there has been growing interest in probiotics as natural, safe, and accessible dietary supplements with the potential to support performance and recovery. Emerging evidence highlights the pivotal role of the gut microbiome in mediating communication along the gut-brain and gut-muscle axes, thereby influencing not only metabolic and immune functions but also neuromuscular adaptation and fatigue resistance. This review explores the mechanisms through which probiotics may enhance exercise performance, mitigate exercise-induced fatigue, and improve physiological adaptation via modulation of inflammation, oxidative stress, and metabolic signaling pathways. Framed within the context of predictive, preventive, and personalized medicine (3P medicine), this paper emphasizes the diagnostic and therapeutic potential of personalized probiotic strategies in optimizing athletic performance through the qualitative and quantitative assessment of microbiota and host responses.
Collapse
Affiliation(s)
- Yongfu Liu
- Department of Rehabilitation, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, 442099, Hubei, China.
- Center for Diabetes Rehabilitation Research, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, 442099, Hubei, China.
| | - Yuting Hu
- Department of Rehabilitation, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan, 442099, Hubei, China
| | - Baolei Ma
- Sports Department of Xi'an Polytechnic University, Xi'an, 710000, Shaanxi, China
| | - Zijun Wang
- College of Medical Technology, Shaanxi University of Chinese Medicine, Xianyan, 712000, Shaanxi, China
| | | |
Collapse
|
11
|
Mahgoup EM. "Gut Microbiota as a Therapeutic Target for Hypertension: Challenges and Insights for Future Clinical Applications" "Gut Microbiota and Hypertension Therapy". Curr Hypertens Rep 2025; 27:14. [PMID: 40261509 DOI: 10.1007/s11906-025-01331-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW Systemic hypertension is a major risk factor for cardiovascular disease and remains challenging to manage despite the widespread use of antihypertensive medications and lifestyle modifications. This review explores the role of gut microbiota in hypertension development and regulation, highlighting key mechanisms such as inflammation, gut-brain axis modulation, and bioactive metabolite production. We also assess the potential of microbiota-targeted therapies for hypertension management. RECENT FINDINGS Emerging evidence indicates that microbial dysbiosis, high-salt diets, and gut-derived metabolites such as short-chain fatty acids (SCFAs) and bile acids significantly influence blood pressure regulation. Preclinical and early clinical studies suggest that interventions targeting gut microbiota, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), and dietary modifications, may help modulate hypertension. However, variability in gut microbiota composition among individuals and limited human trial data pose challenges to translating these findings into clinical practice. While microbiota-based therapies show promise for hypertension management, further research is needed to establish their efficacy and long-term effects. Large-scale, standardized clinical trials are crucial for understanding the therapeutic potential and limitations of gut microbiota interventions. A deeper understanding of the gut-hypertension axis could lead to novel, personalized treatment strategies for hypertension.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt.
- Department of Internal Medicine, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Nurgaziyev M, Kozhakhmetov S, Jarmukhanov Z, Nurgaziyeva A, Sergazy S, Issilbayeva A, Mukhatayev Z, Seidulla S, Sailybayeva A, Bekbossynova M, Kushugulova A. Impact of probiotics and polyphenols on adults with heart failure: a systematic review and meta-analysis. Eur J Med Res 2025; 30:313. [PMID: 40259417 PMCID: PMC12010510 DOI: 10.1186/s40001-025-02538-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/31/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Heart failure poses a significant health concern globally, and despite advancements in treatment, the search for additional, supportive therapeutic options remains crucial. This systematic review and meta-analysis studied the impact of probiotics and polyphenols on heart failure biomarkers, focusing on potential improvements in heart function and inflammation. METHODS We analyzed studies published in Embase, PubMed and Cochrane library from 2012 to 2024, focusing on randomized controlled trials. Our findings are drawn from 5 studies on probiotics, involving 401 participants, and 3 studies on polyphenols with a total of 140 participants. The analysis included assessments of LVEF, hs-CRP, creatinine and NT-proBNP levels in intervention and control groups. RESULTS The probiotics or polyphenols from the included studies did not demonstrate significant changes in the health indicators analyzed for heart failure patients compared to placebo. CONCLUSIONS The systematic review suggested that while the concept of dietary management for heart failure is promising, further research is necessary to validate the efficacy of probiotics and polyphenols as supplementary therapies in heart failure care, by analyzing more diverse health outcomes and patient populations.
Collapse
Affiliation(s)
- Madiyar Nurgaziyev
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan.
| | - Samat Kozhakhmetov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Zharkyn Jarmukhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Ayaulym Nurgaziyeva
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Shynggys Sergazy
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Argul Issilbayeva
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Zhussipbek Mukhatayev
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Symbat Seidulla
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| | - Aliya Sailybayeva
- CF "University Medical Center", Heart Center, Z05G9F9, Astana, Kazakhstan
| | | | - Almagul Kushugulova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Z05H0P9, Astana, Kazakhstan
| |
Collapse
|
13
|
Upadhyay R, Mani S, Sevanan M. Microbiome-based dietary supplements for better development and healthy brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:329-368. [PMID: 40414637 DOI: 10.1016/bs.irn.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Microbiome-based dietary supplements have gained attention for their role in enhancing brain development and cognitive health. The gut microbiome influences neurological functions through the gut-brain axis, impacting neurotransmitter production, immune regulation, and metabolic pathways. Dysbiosis is linked to neurological disorders such as Alzheimer's, Parkinson's, and autism spectrum disorders. This chapter explores dietary interventions targeting the microbiome, emphasising probiotics, prebiotics, and postbiotics. Additionally, AI and machine learning are transforming microbiome research by enabling personalised supplementation strategies tailored to individual gut profiles. Ethical challenges, including data privacy and algorithmic bias, are also discussed. Advances in big data analytics and predictive modelling are paving the way for precision-targeted interventions to optimise brain health. While microbiome-based therapies hold great promise, further clinical validation and regulatory frameworks are needed to ensure their efficacy and accessibility. This chapter highlights the future potential of microbiome-targeted strategies in neuroprotection and cognitive well-being.
Collapse
Affiliation(s)
- Riddhi Upadhyay
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India
| | - Sugumar Mani
- Palamur Biosciences Private Limited, Mahabubnagar, Telangana, India
| | - Murugan Sevanan
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India.
| |
Collapse
|
14
|
Boicean A, Ichim C, Sasu SM, Todor SB. Key Insights into Gut Alterations in Metabolic Syndrome. J Clin Med 2025; 14:2678. [PMID: 40283508 PMCID: PMC12028006 DOI: 10.3390/jcm14082678] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Over time, extensive research has underscored the pivotal role of gut microbiota in the onset and progression of various diseases, with a particular focus on fecal microbiota transplantation (FMT) as a potential therapeutic approach. The practice of transferring fecal matter from a healthy donor to a patient provides valuable insights into how alterations in gut microbiota can impact disease development and how rectifying dysbiosis may offer therapeutic benefits. Re-establishing a balanced symbiotic relationship in the gastrointestinal tract has shown positive results in managing both intestinal and systemic conditions. Currently, one of the most pressing global health issues is metabolic syndrome-a cluster of conditions that includes insulin resistance, lipid imbalances, central obesity and hypertension. In this context, FMT has emerged as a promising strategy for addressing key components of metabolic syndrome, such as improving insulin sensitivity, body weight and lipid profiles. However, further well-structured studies are needed to refine treatment protocols and establish the long-term safety and efficacy of this intervention.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | - Sabina-Maria Sasu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | | |
Collapse
|
15
|
Cedillo-Flores R, Cuevas-Budhart MA, Cavero-Redondo I, Kappes M, Ávila-Díaz M, Paniagua R. Impact of Gut Microbiome Modulation on Uremic Toxin Reduction in Chronic Kidney Disease: A Systematic Review and Network Meta-Analysis. Nutrients 2025; 17:1247. [PMID: 40219004 PMCID: PMC11990722 DOI: 10.3390/nu17071247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Chronic kidney disease is associated with increased intestinal barrier permeability, leading to heightened inflammation and oxidative stress. These changes contribute to complications such as cardiovascular disease, anemia, altered mineral metabolism, and CKD progression. Interventions using prebiotics, probiotics, and synbiotics may mitigate dysbiosis and improve intestinal barrier function, Under this premise, the objective of this network meta-analysis was to evaluate the effect of probiotics, prebiotics, and synbiotics in reducing uremic toxins produced by the gut microbiota in CKD patients. Methods: A systematic review and network meta-analysis of randomized clinical trials (RCTs) was performed in the following databases: Web of Science, Scopus, the Cochrane Register of Controlled Trials, and PubMed published between 2019 and 2023. The analysis focused on the use of prebiotics, probiotics, and synbiotics in CKD patients at stages 3 to 5, as per KDIGO guidelines, and their association with reductions in uremic toxins such as Indoxyl Sulfate, p-Cresyl Sulfate, urea, and creatinine. The risk of bias was assessed using the Cochrane risk of bias tool (RoB 2), with evaluations conducted independently by two reviewers, and a third consulted for disagreements. The study follows the PRISMA statement. Results: The studies included 331 patients, primarily male, across CKD stages 3a to 5. The interventions positively impacted the gut microbiota composition, leading to reductions in free and total p-Cresyl Sulfate (SUCRA: 72.6% and 66.2, respectively) and indoxyl sulfate (SUCRA: 88.5% and 83.1%). Conclusions: The findings suggest that modulating the gut microbiota through these interventions can effectively reduce specific uremic toxins. However, further trials are necessary to better understand microbiota modulation and its impact on intestinal bacterial composition (PROSPERO number: CRD42023438901).
Collapse
Affiliation(s)
- Renata Cedillo-Flores
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (R.C.-F.); (M.Á.-D.)
| | - Miguel Angel Cuevas-Budhart
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (R.C.-F.); (M.Á.-D.)
| | - Iván Cavero-Redondo
- CarVasCare Research Group, Facultad de Enfermería de Cuenca, Universidad de Castilla la Mancha, 16002 Cuenca, Spain;
| | - Maria Kappes
- Faculty of Healthcare Sciencies, Nursing School, Universidad San Sebastián, Puerto Montt 5501842, Chile;
| | - Marcela Ávila-Díaz
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (R.C.-F.); (M.Á.-D.)
| | - Ramón Paniagua
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Centro Médico Nacional Siglo XXI, Ciudad de México 06720, Mexico; (R.C.-F.); (M.Á.-D.)
| |
Collapse
|
16
|
Aleali MS, Mahapatro A, Maddineni G, Paladiya R, Jeanty H, Mohanty E, Mirchandani M, Jahanshahi A, Devulapally P, Alizadehasl A, Tariq MD, Hosseini Jebelli SF, Aliabadi AY, Hashemi SM, Amini-Salehi E. The impact of gut microbiome modulation on anthropometric indices in metabolic syndrome: an umbrella review. Ann Med Surg (Lond) 2025; 87:2263-2277. [PMID: 40212162 PMCID: PMC11981403 DOI: 10.1097/ms9.0000000000003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/25/2025] [Indexed: 04/13/2025] Open
Abstract
Background Metabolic syndrome (MetS) is a complex disorder characterized by a cluster of metabolic risk factors. Recent research highlights the gut microbiome's role in metabolic regulation, suggesting that modulation through probiotics, prebiotics, and synbiotics may provide a novel approach to managing MetS. This umbrella review aims to integrate insights from existing meta-analyses to explore how changes in gut microbiota influence key body measurement indicators in individuals with MetS. Methods A systematic search of PubMed, Scopus, and Web of Science databases identified meta-analyses that assessed the impact of probiotics, prebiotics, or synbiotics on anthropometric indices in MetS patients. Results The results indicated that microbial therapy leads to a significant reduction in body mass index (BMI) (SMD: -0.22; 95% CI: -0.35 to -0.09; P < 0.01) and waist circumference (WC) (SMD: -0.47; 95% CI: -0.80 to -0.15; P < 0.01). However, microbial therapy did not significantly affect body fat mass (SMD: -0.30; 95% CI: -0.64 to 0.02; P = 0.06), body fat percentage (SMD: -0.29; 95% CI: -0.62 to 0.03; P = 0.07), waist-to-hip ratio (SMD: -0.09; 95% CI: -0.46 to 0.28; P = 0.63), and weight (SMD: -0.06; 95% CI: -0.21 to 0.08; P = 0.37). Conclusions Gut microbial modulation, mainly through probiotics and synbiotics, shows promise in reducing BMI and WC in MetS patients. However, its effects on other anthropometric indices remain uncertain, warranting further high-quality research to fully understand microbial interventions' therapeutic potential.
Collapse
Affiliation(s)
- Maryam Sadat Aleali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Ruchir Paladiya
- University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Herby Jeanty
- The Brooklyn Hospital Center, Brooklyn, New York, USA
| | - Elan Mohanty
- Gautam Maddineni, MD Mary Medical Center Apple Valley, Apple Valley, California, USA
| | | | - Ali Jahanshahi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Pavan Devulapally
- Social Determinants of Health Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Azin Alizadehasl
- Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | | | - Seyyed Mohammad Hashemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
17
|
Shah K, Khan AS, Kunwar D, Jacob SR, Akbar A, Singh A, Ahmed MMH. Influence of gut microbiota on the pediatric endocrine system and associated disorders. Ann Med Surg (Lond) 2025; 87:2149-2162. [PMID: 40212169 PMCID: PMC11981368 DOI: 10.1097/ms9.0000000000003099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/15/2025] [Indexed: 04/13/2025] Open
Abstract
The microbiota, a complex assembly of microorganisms residing in various body systems, including the gastrointestinal tract, plays a crucial role in influencing various physiological processes in the human body. The dynamic nature of gut microbiota is especially pronounced in children and is influenced by factors like breastfeeding and antibiotic use. Dysbiosis, characterized by alterations in microbiota composition or function, is associated with several pediatric endocrine disorders, such as precocious puberty, polycystic ovarian syndrome, and diabetes mellitus. This review focuses on the intricate relationship between gut microbiota and the pediatric endocrine system. The aim of this narrative review is to critically examine the existing literature to elucidate the impact of gut microbiota on the pediatric endocrine system and associated disorders. Additionally, potential interventions, such as probiotics and current gaps in knowledge, will be discussed. Despite emerging treatments like probiotics, further research is needed to understand and validate their effectiveness in treating pediatric endocrine disorders associated with dysbiosis.
Collapse
Affiliation(s)
- Krutik Shah
- Byramjee Jeejeebhoy (BJ) Medical College and Civil Hospital, Ahmedabad, India
| | - Alina Sami Khan
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Digbijay Kunwar
- Department of Internal Medicine, Bagahi Primary Healthcare Center, Birgunj, Nepal
| | | | - Anum Akbar
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ajeet Singh
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
18
|
Pereira IM, Pereira M, Lopes JA, Gameiro J. The gut microbiome as a potential therapeutic target in IgA nephropathy. Nephrol Dial Transplant 2025; 40:641-650. [PMID: 39663206 DOI: 10.1093/ndt/gfae274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Indexed: 12/13/2024] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis and a leading cause of kidney failure, with limited treatment options available. The pathophysiology of IgAN remains unclear; however, recent studies suggest that genetic, epigenetic and environmental factors play significant roles. There is also strong evidence linking the gut microbiome to the development of IgAN. In this review, we will examine the relationship between the microbiome and the pathogenesis of IgAN, as well as its potential as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Inês Miguel Pereira
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Marta Pereira
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| | - José António Lopes
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| | - Joana Gameiro
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| |
Collapse
|
19
|
Noor S, Ali S, Summer M, Riaz A, Nazakat L, Aqsa. Therapeutic Role of Probiotics Against Environmental-Induced Hepatotoxicity: Mechanisms, Clinical Perspectives, Limitations, and Future. Probiotics Antimicrob Proteins 2025; 17:516-540. [PMID: 39316257 DOI: 10.1007/s12602-024-10365-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
Hepatotoxicity is one of the biggest health challenges, particularly in the context of liver diseases, often aggravated by gut microbiota dysbiosis. The gut-liver axis has been regarded as a key idea in liver health. It indicates that changes in gut flora caused by various hepatotoxicants, including alcoholism, acetaminophen, carbon tetrachloride, and thioacetamide, can affect the balance of the gut's microflora, which may lead to increased dysbiosis and intestinal permeability. As a result, bacterial endotoxins would eventually enter the bloodstream and liver, causing hepatotoxicity and inducing inflammatory reactions. Many treatments, including liver transplantation and modern drugs, can be used to address these issues. However, because of the many side effects of these approaches, scientists and medical experts are still hoping for a therapeutic approach with fewer side effects and more positive results. Thus, probiotics have become well-known as an adjunctive strategy for managing, preventing, or reducing hepatotoxicity in treating liver injury. By altering the gut microbiota, probiotics offer a secure, non-invasive, and economical way to improve liver health in the treatment of hepatotoxicity. Through various mechanisms such as regulation of gut microbiota, reduction of pathogenic overgrowth, suppression of inflammatory mediators, modification of hepatic lipid metabolism, improvement in the performance of the epithelial barrier of the gut, antioxidative effects, and modulation of mucosal immunity, probiotics play their role in the treatment and prevention of hepatotoxicity. This review highlights the mechanistic effects of probiotics in environmental toxicants-induced hepatotoxicity and current findings on this therapeutic approach's experimental and clinical trials.
Collapse
Affiliation(s)
- Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Anfah Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Laiba Nazakat
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Aqsa
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
20
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
21
|
Cabała S, Herosimczyk A. Diet-Induced Proteomic and Metabolomic Signatures in Chronic Kidney Disease: A Precision Nutrition Approach. Metabolites 2025; 15:211. [PMID: 40137175 PMCID: PMC11943711 DOI: 10.3390/metabo15030211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Diet is a key modifiable factor that can either support renal health or accelerate the onset and progression of chronic kidney disease (CKD). Recent advances in multiomics, particularly proteomics and metabolomics, significantly enhanced our understanding of the molecular mechanisms linking diet to CKD risk. Proteomics offers a comprehensive analysis of protein expression, structure, and interactions, revealing how dietary components regulate cellular processes and signaling pathways. Meanwhile, metabolomics provides a detailed profile of low-molecular-weight compounds, including endogenous metabolites and diet-derived molecules, offering insights into the metabolic states that influence kidney function. Methods: We have conducted a narrative review of key papers from databases such as PubMed, Scopus, and Web of Science to explore the potential of proteomic and metabolomic analysis in identifying molecular signatures associated with diet in human and animal biological samples, such as blood plasma, urine, and in kidney tissues. These signatures help elucidate how specific foods, food groups, and overall dietary patterns may either contribute to or mitigate CKD risk. Results: Recent studies the impact of high-fat diets on protein expression involved in energy metabolism, inflammation, and fibrosis, identifying early biomarkers of kidney injury. Metabolic, including disruptions in in fatty acid metabolism, glucose regulation, and amino acid pathways, have been recognized as key indicators of CKD risk. Additionally, several studies explore specific metabolites found in biological fluids and renal tissue in response to protein-rich foods, assessing their potential roles in a progressive loss of kidney function. Emerging evidence also suggests that dietary interventions targeting the gut microbiota may help alleviate inflammation, oxidative stress, and toxin accumulation in chronic kidney disease. Notably, recent findings highlight metabolomic signatures linked to beneficial shifts in gut microbial metabolism, particularly in the context of prebiotic supplementation. Conclusions: By integrating proteomics and metabolomics, future research can refine precision nutrition strategies, helping mitigate CKD progression. Expanding large-scale studies and clinical trials will be essential in translating these molecular insights into actionable dietary guidelines.
Collapse
Affiliation(s)
| | - Agnieszka Herosimczyk
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland;
| |
Collapse
|
22
|
Jha SS, Jeyaraman N, Jeyaraman M, Ramasubramanian S, Muthu S, Santos GS, da Fonseca LF, Lana JF. Cross-talks between osteoporosis and gut microbiome. World J Orthop 2025; 16:102274. [PMID: 40124724 PMCID: PMC11924030 DOI: 10.5312/wjo.v16.i3.102274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 03/12/2025] Open
Abstract
The gut microbiome comprises a vast community of microbes inhabiting the human alimentary canal, playing a crucial role in various physiological functions. These microbes generally live in harmony with the host; however, when dysbiosis occurs, it can contribute to the pathogenesis of diseases, including osteoporosis. Osteoporosis, a systemic skeletal disease characterized by reduced bone mass and increased fracture risk, has attracted significant research attention concerning the role of gut microbes in its development. Advances in molecular biology have highlighted the influence of gut microbiota on osteoporosis through mechanisms involving immunoregulation, modulation of the gut-brain axis, and regulation of the intestinal barrier and nutrient absorption. These microbes can enhance bone mass by inhibiting osteoclast differentiation, inducing apoptosis, reducing bone resorption, and promoting osteoblast proliferation and maturation. Despite these promising findings, the therapeutic effectiveness of targeting gut microbes in osteoporosis requires further investigation. Notably, gut microbiota has been increasingly studied for their potential in early diagnosis, intervention, and as an adjunct therapy for osteoporosis, suggesting a growing utility in improving bone health. Further research is essential to fully elucidate the therapeutic potential and clinical application of gut microbiome modulation in the management of osteoporosis.
Collapse
Affiliation(s)
- Shiva Shankar Jha
- Department of Orthopaedics, Harishchandra Orthopaedic Research Institute, Patna 880023, Bihar, India
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
23
|
Menozzi E, Schapira AHV, Borghammer P. The Gut-Brain Axis in Parkinson disease: Emerging Concepts and Therapeutic Implications. Mov Disord Clin Pract 2025. [PMID: 40079755 DOI: 10.1002/mdc3.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The gut-brain axis, i.e. the bidirectional communication system between the gut and the brain, has become of central importance in Parkinson disease (PD) research over the past 20 years. AIMS We aimed to describe the milestones of the gut-brain axis research in PD and the development of theories proposing the involvement of the gastrointestinal tract in PD pathogenesis. METHODS We searched PubMed using the terms 'gut-brain axis' AND 'Parkinson disease', and selected relevant articles to provide the foundation for reconstructing an historical overview of the gut-brain axis research in PD. RESULTS Mounting evidence from preclinical, clinical and post-mortem studies suggests that a subgroup of PD patients present with a range of prodromal symptoms (e.g., autonomic dysfunction, rapid eye movement sleep behaviour disorder) which reflect initial accumulation and later spread of pathological α-synuclein rostrally from the gastrointestinal tract ("body-first" PD). Through neural connections along the gut-brain axis, pathological α-synuclein may spread to the brain, producing clinically manifest disease. Recently, two mechanisms involving the gut-brain axis have attracted increasing attention for their role in PD pathogenesis and progression, namely the perturbation of the composition of the microorganisms living in the gut (the gut microbiome), and the dysfunction of enteroendocrine cells. CONCLUSION Treatments targeting the gut-brain axis, especially the gut microbiome and the enteroendocrine cells pathway, could potentially slow disease progression or even prevent disease onset. Among these, pre/probiotics, faecal microbiota transplantation, and glucagon-like peptide-1 receptor agonists, have entered advanced stages of clinical trials in humans and shown potential symptomatic and disease-modifying effects.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
24
|
Tain YL, Lin YJ, Hsu CN. Breastfeeding and Future Cardiovascular, Kidney, and Metabolic Health-A Narrative Review. Nutrients 2025; 17:995. [PMID: 40290039 PMCID: PMC11944316 DOI: 10.3390/nu17060995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The benefits of breastfeeding for both mother and infant are generally recognized; however, the connections between breast milk, lactation, and long-term offspring health and disease remain incompletely understood. Cardiovascular-kidney-metabolic syndrome (CKMS) has become a major global public health challenge. Insufficient breast milk supply, combined with various early-life environmental factors, markedly increases the future risk of CKMS, as highlighted by the developmental origins of health and disease (DOHaD) concept. Given its richness in nutrients and bioactive components essential for infant health, this review focuses on reprogramming strategies involving breast milk to improve offspring's cardiovascular, kidney, and metabolic health. It also highlights recent experimental advances in understanding the mechanisms driving CKMS programming. Cumulatively, the evidence suggests that lactational impairment heightens the risk of CKMS development. In contrast, early interventions during the lactation period focused on animal models that leverage breast milk components in response to early-life cues show potential in improving cardiovascular, kidney, and metabolic outcomes-an area warranting further investigation and clinical translation.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Ying-Jui Lin
- Division of Critical Care, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Division of Cardiology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Department of Early Childhood Care and Education, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
25
|
Zandifar A, Badrfam R, Mohammaditabar M, Kargar B, Goodarzi S, Hajialigol A, Ketabforoush S, Heidari A, Fathi H, Shafiee A, Pourjafar H. The Effect of Prebiotics and Probiotics on Levels of Depression, Anxiety, and Cognitive Function: A Meta-Analysis of Randomized Clinical Trials. Brain Behav 2025; 15:e70401. [PMID: 40038860 PMCID: PMC11879892 DOI: 10.1002/brb3.70401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 03/06/2025] Open
Abstract
INTRODUCTION Recent studies have emphasized the relationship between mental health and the human intestine microbiota. In this study, we evaluate the effect of consuming Biotics, on levels of depression, anxiety, and cognitive function. METHODS This meta-analysis adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standards. We searched MEDLINE (PubMed), Cochrane Library, Scopus, Web of Science, and ClinicalTrials.gov. All full-text articles and major reviews were manually searched for additional studies. RESULTS The initial analysis was based on the concept that consuming Biotics causes changes in anxiety, measured using various instruments. This analysis showed that consuming Biotics significantly reduced anxiety in our study participants (SMD = 0.2894, Z = 2.46, P = 0.0139, I^2 = 92.4%). The meta-analysis included 4295 samples (2194 in the experimental group and 2101 in the control group). In terms of depression, the analysis showed that consuming Biotics significantly reduced depression in our study participants (SMD = 0.2942, Z = 2.13, P = 0.0335, I^2 = 91.7%). The meta-analysis included 3179 samples (1603 in the experimental group and 1576 in the control group). Regarding cognitive function, the analysis showed that consuming Biotics significantly improved cognitive function in our study participants (SMD = 0.4819, Z = 3.00, P = 0.0027, I^2 = 77.9%). The meta-analysis included 915 samples (470 in the experimental group and 445 in the control group). CONCLUSIONS Our results indicate that most recent studies support the effectiveness of probiotics in reducing symptoms of anxiety, depression, and cognitive issues despite some discrepancies in the findings. People with mild symptoms may experience greater benefits from taking probiotics. TRIAL REGISTRATION PROSPERO registration ID: CRD42024589507.
Collapse
Affiliation(s)
- Atefeh Zandifar
- Dietary Supplements and Probiotic Research CenterAlborz University of Medical SciencesKarajIran
- Clinical Research Development Unit of Imam Hossein Medical Education CenterAlborz University of Medical SciencesKarajIran
- Social Determinants of Health Research CenterAlborz University of Medical SciencesKarajIran
| | - Rahim Badrfam
- Department of Psychosomatic MedicineShariati Hospital, Alborz University of Medical SciencesKarajAlborzIran
- Non‐communicable Diseases Research CenterAlborz University of Medical SciencesKarajAlborzIran
- Community Mental Health CenterAlborz University of Medical SciencesKarajAlborzIran
| | - Mahdi Mohammaditabar
- Student Research Committee, School of MedicineAlborz University of Medical SciencesKarajIran
- Alborz Office of Universal Scientific Education and Research Network (USERN)Alborz University of Medical SciencesKarajIran
| | - Bita Kargar
- Tehran Medical Sciences Islamic Azad UniversityTehranIran
| | - Saba Goodarzi
- Student Research Committee, School of MedicineAlborz University of Medical SciencesKarajIran
| | - Amirhossein Hajialigol
- Alborz Office of Universal Scientific Education and Research Network (USERN)Alborz University of Medical SciencesKarajIran
| | - Shera Ketabforoush
- Student Research CommitteeTehran Medical Sciences Islamic Azad UniversityTehranIran
| | - Afshin Heidari
- School of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Hanie Fathi
- Student Research Committee, School of MedicineAlborz University of Medical SciencesKarajIran
| | - Arman Shafiee
- Student Research Committee, School of MedicineAlborz University of Medical SciencesKarajIran
| | - Hadi Pourjafar
- Dietary Supplements and Probiotic Research CenterAlborz University of Medical SciencesKarajIran
| |
Collapse
|
26
|
Khouma M, Diallo M, Sow D, Allen S, Faye B. Efficacy of prebiotic, probiotic, and synbiotics in improving growth in children under age five years in Africa: A protocol for a systematic review. Nutr Health 2025; 31:23-29. [PMID: 39370911 DOI: 10.1177/02601060241282357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background: Stunting is among the main obstacles to human development affecting millions of children worldwide, particularly in the sub-Saharan Africa region. Randomized clinical trials have shown the positive effects of prebiotics, probiotics, and synbiotics in improving growth in children and toddlers. However, although the global mobilization to tackle its challenges in their different aspects is visible, it remains to define effective large-scale up interventions and strategies to obtain long-lasting impacts. Objective: The objective of this review is to re-evaluate the efficacy of prebiotics, probiotics, and/or synbiotics on growth in children 0 to 5 years in Africa including recently published studies. Methods: Systematic search will be carried out in Pubmed, Science Direct, clinicaltrial.org, and Google Scholar. Both randomized and observational studies that assess the association between prebiotics, probiotics, and synbiotics, and health benefits and growth in children under 5 years of age will be included in the review. PRISMA-P (preferred reporting items for systematic review and meta-analysis protocols) will be used used for this protocol, and PRISMA will be used for the systematic review. The Cochrane Risk Assessment Tool will be used to assess the quality of eligible studies. If the compiled data are appropriate and sufficient enough, we will perform a meta-analysis using RevMan software. Conclusion: This review will provide up-to-date and reliable information on the efficacy of prebiotics, probiotics, and synbiotics on the growth of children under 5 years of age especially in developing countries. PROSPERO registration number CRD42022343138.
Collapse
Affiliation(s)
- Marietou Khouma
- Department of Animal Biology, Faculty of Science and Technology, Cheikh Anta Diop University Dakar, Dakar, Senegal
| | - Mamadou Diallo
- Department of Animal Biology, Faculty of Science and Technology, Cheikh Anta Diop University Dakar, Dakar, Senegal
| | - Doudou Sow
- Service of Parasitology-Mycologie, UFR Health Science, University Gaston Berger of Saint Louis, Saint-Louis, Senegal
| | - Steve Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Babacar Faye
- Faculty of Medicine, Pharmacy and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| |
Collapse
|
27
|
Zalila-Kolsi I, Dhieb D, Osman HA, Mekideche H. The Gut Microbiota and Colorectal Cancer: Understanding the Link and Exploring Therapeutic Interventions. BIOLOGY 2025; 14:251. [PMID: 40136508 PMCID: PMC11939563 DOI: 10.3390/biology14030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
CRC remains a significant public health challenge due to its high prevalence and mortality rates. Emerging evidence highlights the critical role of the gut microbiota in both the pathogenesis of CRC and the efficacy of treatment strategies, including chemotherapy and immunotherapy. Dysbiosis, characterized by imbalances in microbial communities, has been implicated in CRC progression and therapeutic outcomes. This review examines the intricate relationship between gut microbiota composition and CRC, emphasizing the potential for microbial profiles to serve as biomarkers for early detection and prognosis. Various interventions, such as prebiotics, probiotics, postbiotics, fecal microbiota transplantation, and dietary modifications, aim to restore microbiota balance and shift dysbiosis toward eubiosis, thereby improving health outcomes. Additionally, the integration of microbial profiling into clinical practice could enhance diagnostic capabilities and personalize treatment strategies, advancing the field of oncology. The study of intratumoral microbiota offers new diagnostic and prognostic tools that, combined with artificial intelligence algorithms, could predict treatment responses and assess the risk of adverse effects. Given the growing understanding of the gut microbiome-cancer axis, developing microbiota-oriented strategies for CRC prevention and treatment holds promise for improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Imen Zalila-Kolsi
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Dhoha Dhieb
- College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussam A. Osman
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Hadjer Mekideche
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| |
Collapse
|
28
|
Khouma M, Diallo M, Sow D, Djigal A, Diop NS, Allen S, Faye B. Efficacy of prebiotic, probiotic and synbiotic administration in improving growth in children aged 0-59 months living in low- and middle-income countries: a systematic review and meta-analysis. BMC Pediatr 2025; 25:149. [PMID: 40011839 PMCID: PMC11866818 DOI: 10.1186/s12887-025-05503-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Poor growth is one of the major obstacles to human development, affecting millions of children under the age of 5 years, particularly those living in low- and middle-income countries (LMICs). The objective of this review was to evaluate the efficacy of administering pre-, pro- or synbiotics on the growth of children aged 0-59 months living in LMICs. METHODS Google scholar, Pubmed, clinical trial.org and Science Direct databases were searched in April 2023 for randomised controlled trials of pre-, pro- or synbiotics that evaluated growth in under fives in LMICs. The primary outcome were weight and height gain. Secondary outcomes were head circumference, body mass index gain and Z score. Random-effects meta-analysis was used to calculate mean differences for continuous outcomes. Grading of Recommendations Assessment, Development and Evaluation criteria was used to assess certainty of the evidence. RESULTS Eight trials involving 1375 children under 5 years of age were identified. Meta-analysis of 6 RCTs (n = 991 children) revealed a significant difference in favor of the experimental group (n = 579) compared the control group (n = 412) for weight gain: (MD = 0.33 kg, 95% CI 0.11 to 0.55); low-certainty evidence. Sub-group analysis revealed that pre-, pro, or synbiotics may be more effective in malnourished that healthy children (p = 0.003). Meta-analysis of height gain for 4 RCTs (n = 845) found that there was no significant difference between the experimental group (n = 496) and the control group (n = 349) (MD = 0.31 cm; 95% CI -0.36 to 0.98); low-certainty evidence. In sub-group analysis, prebiotics had a greater impact on height gain than synbiotics (p = 0.03). In the only study reporting an increase in head circumference (n = 32 children), this was not improved by the administration of synbiotics. However, administration of synbiotics to undernourished children significantly improved BMI gain. CONCLUSION The evidence for the administration of pre-, pro- or synbiotics on the growth of children in LMICs is weak. Administration of pre-, pro- or synbiotics may improve weight gain in both healthy and malnourished children. Prebiotics and synbiotics had a significant effect on weight gain. Further research is needed due to the small number of studies, short duration of administration and small sample size.
Collapse
Affiliation(s)
- Marietou Khouma
- Department of Animal Biology, Faculty of Science and Technology, Cheikh Anta Diop University of Dakar, Dakar, Senegal.
| | - Mamadou Diallo
- Department of Animal Biology, Faculty of Science and Technology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Doudou Sow
- Faculty Parasitology-Mycologie, UFR Sciences de La Santé, Gaston Berger University of Saint Louis, Saint Louis, Senegal
| | - Aicha Djigal
- Department of Animal Biology, Faculty of Science and Technology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Ndeye Sokhna Diop
- Faculty of Medecine, Pharmacy and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Stephen Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Babacar Faye
- Faculty of Medecine, Pharmacy and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| |
Collapse
|
29
|
Charitos IA, Scacco S, Cotoia A, Castellaneta F, Castellana G, Pasqualotto F, Venneri M, Ferrulli A, Aliani M, Santacroce L, Carone M. Intestinal Microbiota Dysbiosis Role and Bacterial Translocation as a Factor for Septic Risk. Int J Mol Sci 2025; 26:2028. [PMID: 40076650 PMCID: PMC11900423 DOI: 10.3390/ijms26052028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
The human immune system is closely linked to microbiota such as a complex symbiotic relationship during the coevolution of vertebrates and microorganisms. The transfer of microorganisms from the mother's microbiota to the newborn begins before birth during gestation and is considered the initial phase of the intestinal microbiota (IM). The gut is an important site where microorganisms can establish colonies. The IM contains polymicrobial communities, which show complex interactions with diet and host immunity. The tendency towards dysbiosis of the intestinal microbiota is influenced by local but also extra-intestinal factors such as inflammatory processes, infections, or a septic state that can aggravate it. Pathogens could trigger an immune response, such as proinflammatory responses. In addition, changes in the host immune system also influence the intestinal community and structure with additional translocation of pathogenic and non-pathogenic bacteria. Finally, local intestinal inflammation has been found to be an important factor in the growth of pathogenic microorganisms, particularly in its role in sepsis. The aim of this article is to be able to detect the current knowledge of the mechanisms that can lead to dysbiosis of the intestinal microbiota and that can cause bacterial translocation with a risk of infection or septic state and vice versa.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Doctoral School, Applied Neurosciences, University of Bari (UNIBA), 70124 Bari, Italy
| | - Salvatore Scacco
- Dipartimento di Biomedicina Traslazionale e Neuroscienze (DiBraiN), Scuola di Medicina, Università Degli Studi di Bari, Aldo Moro, 70124 Bari, Italy;
- U.O. Medicina, Ospedale Mater Dei-CBH, 70125 Bari, Italy
| | - Antonella Cotoia
- Department of Intensive Care, University Hospital of Foggia, 71121 Foggia, Italy
| | - Francesca Castellaneta
- U.O.C. Servizio di Immunoematologia e Medicina Trasfusionale—S.I.M.T. Ospedale Di Venere, 70131 Bari, Italy;
| | - Giorgio Castellana
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Federico Pasqualotto
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Department of Public Health and Infectious Diseases, Pulmonary Division, Sapienza University of Rome, Policlinico Umberto I Hospital, Rome, Via del Policlinico 155, 00155 Rome, Italy
| | - Maria Venneri
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Angela Ferrulli
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Maria Aliani
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, The University of Bari, 70124 Bari, Italy;
| | - Mauro Carone
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| |
Collapse
|
30
|
Naveed M, Khan S, Aziz T, Ur Rehman S, Makhdoom SI, Al-Harbi M, Alshammari A. Elucidation of Bacterial-Mediated Hesperidin Transformation, Structural Evaluation, and Computational Drug Targeting against Helicobacter pylori. Mol Biotechnol 2025:10.1007/s12033-025-01406-8. [PMID: 39992485 DOI: 10.1007/s12033-025-01406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Biotransformation, a dynamic process conducted by microorganisms, holds promise in modifying natural compounds for enhanced therapeutic potential. In this study, we leverage bacterial systems to catalyze the transformation of hesperidin, obtained from Citrus sinensis, aiming for structural modifications that could optimize its bioactivity and computational targeting against Helicobacter pylori. Multiple bacterial species were employed to perform biotransformation reactions. HPLC and FTIR analyses were used to determine structural modifications and bio-transformed products. The reaction in which hesperidin metabolite was formed was catalyzed by Bacillus spp. The transformed products, along with the original compound, underwent rigorous bioactivity testing to evaluate their potential in combating oxidative stress, inflammation, and diabetes. Employing well-established in vitro methods, we assessed the bio-transformed derivatives for antioxidant efficacy, revealing an impressive 94% inhibition of free radicals compared to hesperidin. In terms of anti-inflammatory activity, the results showcased a substantial 92% inhibition, while the assessment of antidiabetic activity demonstrated a notable 85% inhibition. The hesperidin metabolites were more active than hesperidin in biological activity evaluations. So, the bio-transformed compound derived from hesperidin, along with pure compound, was used to design a computational drug targeting the bacterium H. pylori. Among these two compounds, the bio-transformed product of hesperidin with an alkyl amine exhibited the highest docking energy of - 180.26 kJ/mol, as compared to pure compound. SwissADME provided valuable insights into the compound's drug-likeness like 0.55 bioavailability and 8.41 synthetic accessibility. ProTox-II evaluated different toxicity endpoints with a 0.96 probability of being inactive in cytotoxicity. These findings support the potential of the bio-transformed compound as a promising candidate for further investigation and development as a drug against H. pylori.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan.
| | - Sara Khan
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina Arta, Ioannina, 47132, Greece.
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54792, Punjab, Pakistan.
| | - Shafique Ur Rehman
- Department of Basic and Applied Chemistry, Faculty of Sciences, University of Central Punjab, Lahore, 54590, Pakistan
| | - Syeda Izma Makhdoom
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Mitub Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
31
|
Sa'aid N, Tan JS. From probiotic fermentation to functional drinks: a review on fruit juices with lactic acid bacteria and prebiotics. Prep Biochem Biotechnol 2025:1-20. [PMID: 39968919 DOI: 10.1080/10826068.2025.2467441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
In recent years, the demand for probiotic beverages has surged, with dairy products traditionally serving as the primary sources of probiotics. However, many consumers face health issues such as lactose intolerance, milk allergies, and high cholesterol, which prevent them from consuming dairy products. This has led to the exploration of nondairy alternatives, particularly fruit juices, as carriers for probiotics. Lactic acid bacteria (LAB) have been identified as beneficial probiotics that can be incorporated into these beverages. The inclusion of prebiotics, such as inulin and galacto-oligosaccharides (GOS), in fruit juices has shown promise in enhancing the growth and activity of LAB, thereby creating functional beverages that support digestive health. Despite numerous studies on fruit juice fermentation, there is limited data on the optimal pairing of probiotics and prebiotics to develop stable, nondairy functional drinks. This review underscores the potential of lactic acid fermentation and the integration of prebiotics and probiotics in fruit juices, highlighting the necessity for further research to optimize these combinations for enhanced health benefits and improved beverage stability.
Collapse
Affiliation(s)
- Nurhazwani Sa'aid
- School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Pulau Pinang, Malaysia
| | - Joo Shun Tan
- School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Pulau Pinang, Malaysia
| |
Collapse
|
32
|
Farah A, Paul P, Khan AS, Sarkar A, Laws S, Chaari A. Targeting gut microbiota dysbiosis in inflammatory bowel disease: a systematic review of current evidence. Front Med (Lausanne) 2025; 12:1435030. [PMID: 40041456 PMCID: PMC11876558 DOI: 10.3389/fmed.2025.1435030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction The dysbiosis of the gut microbiota has been identified as a central factor in the pathogenesis of inflammatory bowel disease (IBD), a chronic condition characterized by frequent recurrence and various adverse effects of traditional therapies. While treatments targeting the gut microbiota show promise, their efficacy in IBD management still requires extensive evaluation. Our systematic review analyzes recent studies to elucidate the advancements and challenges in treating IBD using microbial-based therapies. Methods Through a comprehensive systematic review spanning key scientific databases-PubMed, Embase, Cochrane, Web of Science, Scopus, and Google Scholar-we scrutinized the impact of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) on individuals with IBD. Our detailed analysis covered study and participant demographics, along with seven key outcome measures: disease activity index, inflammatory markers, serum cytokines, microbiome composition, adverse effects, and the rates of remission and relapse. Results From 6,080 initial search hits, we included 71 studies that assessed various interventions compared to placebo or standard medical therapy. Although there was notable variation in clinical results while assessing different outcomes, overall, probiotics, prebiotics, and synbiotics enhanced the success rates in inducing remission among IBD patients. Furthermore, we noted significant reductions in levels of pro-inflammatory markers and cytokines. Additionally, the requirement for steroids, hospitalization, and poor outcomes in endoscopic and histological scores were significantly reduced in individuals undergoing FMT. Conclusion Our investigation highlights the potential of targeting gut microbiota dysbiosis with microbial-based therapies in patients with IBD. We recommend conducting larger, placebo-controlled randomized trials with extended follow-up periods to thoroughly assess these treatments' clinical efficacy and safety before widespread recommendations for clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha, Qatar
| |
Collapse
|
33
|
Kumar S, Mukherjee R, Gaur P, Leal É, Lyu X, Ahmad S, Puri P, Chang CM, Raj VS, Pandey RP. Unveiling roles of beneficial gut bacteria and optimal diets for health. Front Microbiol 2025; 16:1527755. [PMID: 40041870 PMCID: PMC11877911 DOI: 10.3389/fmicb.2025.1527755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome plays a pivotal role in human health, influencing digestion, immunity, and disease prevention. Beneficial gut bacteria such as Akkermansia muciniphila, Adlercreutzia equolifaciens, and Christensenella minuta contribute to metabolic regulation and immune support through bioactive metabolites like short-chain fatty acids (SCFAs). Dietary patterns rich in prebiotics, fermented foods, and plant-based bioactive compounds, including polyphenols and flavonoids, promote microbiome diversity and stability. However, challenges such as individual variability, bioavailability, dietary adherence, and the dynamic nature of the gut microbiota remain significant. This review synthesizes current insights into gut bacteria's role in health, emphasizing the mechanisms by which dietary interventions modulate microbiota. Additionally, it highlights advancements in microbiome-targeted therapies and the transformative potential of personalized nutrition, leveraging microbiota profiling and artificial intelligence (AI) to develop tailored dietary strategies for optimizing gut health and mitigating chronic inflammatory disorders. Addressing these challenges requires a multidisciplinary approach that integrates scientific innovation, ethical frameworks, and practical implementation strategies.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Pratibha Gaur
- Centre for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
| | - Élcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha'il, Saudi Arabia
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - V. Samuel Raj
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Ramendra Pati Pandey
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| |
Collapse
|
34
|
Tain YL, Lin YJ, Hsu CN. Animal Models for Studying Developmental Origins of Cardiovascular-Kidney-Metabolic Syndrome. Biomedicines 2025; 13:452. [PMID: 40002865 PMCID: PMC11853432 DOI: 10.3390/biomedicines13020452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular-kidney-metabolic syndrome (CKMS) has become a significant global health challenge. Since CKMS often originates early in life, as outlined by the developmental origins of health and disease (DOHaD) concept, prevention is a more effective strategy than treatment. Various animal models, classified by environmental exposures or mechanisms, are used to explore the developmental origins of CKMS. However, no single model can fully replicate all aspects of CKMS or its clinical stages, limiting the advancement of preventive and therapeutic strategies. This review aims to assist researchers by comparing the strengths and limitations of common animal models used in CKMS programming studies and highlighting key considerations for selecting suitable models.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Ying-Jui Lin
- Division of Critical Care, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Division of Cardiology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Department of Early Childhood Care and Education, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Depatrtment of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| |
Collapse
|
35
|
Rui X, Fu K, Wang H, Pan T, Wang W. Formation Mechanisms of Protein Coronas on Food-Related Nanoparticles: Their Impact on Digestive System and Bioactive Compound Delivery. Foods 2025; 14:512. [PMID: 39942105 PMCID: PMC11817056 DOI: 10.3390/foods14030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
The rapid development of nanotechnology provides new approaches to manufacturing food-related nanoparticles in various food industries, including food formulation, functional foods, food packaging, and food quality control. Once ingested, nanoparticles will immediately adsorb proteins in the biological fluids, forming a corona around them. Protein coronas alter the properties of nanoparticles, including their toxicity, cellular uptake, and targeting characteristics, by altering the aggregation state. In addition, the conformation and function of proteins and enzymes are also influenced by the formation of protein coronas, affecting the digestion of food products. Since the inevitable application of nanoparticles in food industries and their subsequent digestion, a comprehensive understanding of protein coronas is essential. This systematic review introduces nanoparticles in food and explains the formation of protein coronas, with interactions between proteins and nanoparticles. Furthermore, the potential origin of nanoparticles in food that migrate from packaging materials and their fates in the gastrointestinal tract has been reviewed. Finally, this review explores the possible effects of protein coronas on bioactive compounds, including probiotics and prebiotics. Understanding the formation mechanisms of protein coronas is crucial, as it enables the design of tailored delivery systems to optimize the bioavailability of bioactive compounds.
Collapse
Affiliation(s)
| | | | | | | | - Wenjun Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
36
|
Chudzicka-Strugała I, Kubiak A, Banaszewska B, Wysocka E, Zwozdziak B, Siakowska M, Pawelczyk L, Duleba AJ. Six-month randomized, placebo controlled trial of synbiotic supplementation in women with polycystic ovary syndrome undergoing lifestyle modifications. Arch Gynecol Obstet 2025; 311:499-506. [PMID: 39636391 PMCID: PMC11890239 DOI: 10.1007/s00404-024-07833-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE To determine whether long-term administration of synbiotics affects clinical, endocrine and metabolic aspects of polycystic ovary syndrome (PCOS) in overweight and obese subjects undergoing intensive lifestyle modifications. METHODS During six-month trial, all subjects underwent intensive lifestyle modifications (diet and exercise). The subjects were randomized (1:1) to receive synbiotic supplementation (Synbiotic Group) or placebo (Placebo Group). RESULTS Subjects in the Placebo Group and the Synbiotic Group experienced significant reduction of BMI (- 8% and - 11%, respectively; both at P < 0.0001) and body fat percentage (- 11% and - 14%, respectively; both at P < 0.0001). These effects were statistically comparable for both groups. Total testosterone was not significantly changed in the Placebo Group (- 5%, P = 0.41) while it greatly declined in the Synbiotic Group (- 40%; P < 0.0001); the difference between these groups was significant (P = 0.0002). Synbiotic supplementation was superior to placebo in reducing LH (- 21%; P = 0.047), total cholesterol (- 6%; P = 0.002), low-density lipoprotein cholesterol (- 6%; P = 0.044), triglycerides (- 29%; P = 0.049), LPS (- 23%; P = 0.001) and LPS-binding protein (- 21%; P = 0.001). CONCLUSIONS Synbiotic supplementation led to a marked improvement of several key clinical and laboratory aspects of PCOS including an improvement of hyperandrogenism, lipid profile, and markers of endotoxemia. TRIAL REGISTRATION Clinical Trial Registration Number: NCT03325023 (URL, clinicaltrials.gov; date of registration 10/26/2017).
Collapse
Affiliation(s)
| | - Anna Kubiak
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 60-535, Poznan, Poland
| | - Beata Banaszewska
- Department of Laboratory Diagnostics, Poznan University of Medical Sciences, 60-569, Poznan, Poland
| | - Ewa Wysocka
- Department of Laboratory Diagnostics, Poznan University of Medical Sciences, 60-569, Poznan, Poland
| | - Barbara Zwozdziak
- Department of Medical Microbiology, Poznan University of Medical Sciences, 60-535, Poznan, Poland
| | - Martyna Siakowska
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 60-535, Poznan, Poland
| | - Leszek Pawelczyk
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 60-535, Poznan, Poland
| | - Antoni J Duleba
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, 9500 Gilman Drive, 0633, San Diego, CA, 92093-0633, USA.
| |
Collapse
|
37
|
Medina JGS, Camacho JLC, Ruiz Garcia J, Mira A, Martínez Martínez RE, Comas-García M, Rangel AG, Pozos-Guillén A, Romo SA. Streptococcus dentisani inhibits the growth of Candida albicans and Candida glabrata: in vitro assay. Int Microbiol 2025; 28:289-298. [PMID: 38844735 DOI: 10.1007/s10123-024-00525-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Probiotic bacteria inhibit aggregation, biofilm formation, and dimorphism of Candida spp. However, the effects of a new probiotic, Streptococcus dentisani, on the growth of Candida albicans and Candida glabrata biofilms are unknown. OBJECTIVE To determine the effect of S. dentisani on the different phases of C. albicans and C. glabrata biofilm development. METHODS Growth quantification and ultrastructural analyses were performed on biofilms of C. albicans ATCC 90028, C. glabrata ATCC 2001, and clinical isolates of C. albicans from oral candidiasis (CA-C1), caries (CA-CR1), and periodontal pocket (CA-P1) treated with cell suspensions of S. dentisani CECT 7746. Cell viability was determined by quantifying colony-forming units (CFU/mL). The ultrastructural analyses were done with atomic force microscopy. RESULTS S. dentisani induced a significant reduction (p < 0.05) of CFU/mL of immature and mature biofilm in all strains of C. albicans and C. glabrata. Microscopic analysis revealed that S. dentisani reduced C. albicans density in mixed biofilm. The fungus-bacteria interaction affected cell membrane integrity in yeast. CONCLUSION For the first time, our data elucidate the antifungal effect of S. dentisani on the development of C. albicans and C. glabrata biofilms, supporting its usefulness as a niche-specific probiotic to prevent and treat oral dysbiosis.
Collapse
Affiliation(s)
- Juan Gerardo Sánchez Medina
- Department of Microbiology, Faculty of Dentistry, Autonomous University of San Luis Potosi, Manuel Nava No. 2, Col Universitaria, 78290, San Luis Potosí, S.L.P., Mexico
| | | | - Jaime Ruiz Garcia
- Laboratory of Biological Physics, Physics Institute, Autonomous University of San Luis Potosí, San Luis Potosí, México
| | - Alex Mira
- Department of Health and Genomics, Oral Microbiome Lab, FISABIO Foundation, Valencia, Spain
| | - Rita Elizabeth Martínez Martínez
- Periodontics Postgraduate Program, Faculty of Dentistry, Autonomous University of San Luis Potosi, San Luis Potosí, S.L.P., Mexico
| | - Mauricio Comas-García
- Department of Sciences, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosi, San Luis Potosí, S.L.P., Mexico
| | - Arturo Garrocho Rangel
- Periodontics Postgraduate Program, Faculty of Dentistry, Autonomous University of San Luis Potosi, San Luis Potosí, S.L.P., Mexico
| | - Amaury Pozos-Guillén
- Basic Sciences Laboratory, Faculty of Dentistry, Autonomous University of San Luis Potosí, San Luis Potosí, S.L.P., Mexico
| | - Saray Aranda Romo
- Department of Microbiology, Faculty of Dentistry, Autonomous University of San Luis Potosi, Manuel Nava No. 2, Col Universitaria, 78290, San Luis Potosí, S.L.P., Mexico.
- Periodontics Postgraduate Program, Faculty of Dentistry, Autonomous University of San Luis Potosi, San Luis Potosí, S.L.P., Mexico.
- Diagnostic Clinic Dentistry School, Autonomous University of San Luis Potosí, San Luis Potosí, S.L.P., Mexico.
| |
Collapse
|
38
|
Kiruthika K, Suganthi A, Johnson Thangaraj Edward YS, Anandham R, Renukadevi P, Murugan M, Bimal Kumar Sahoo, Mohammad Ikram, Kavitha PG, Jayakanthan M. Role of Lactic Acid Bacteria in Insecticide Residue Degradation. Probiotics Antimicrob Proteins 2025; 17:81-102. [PMID: 38819541 DOI: 10.1007/s12602-024-10298-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
Lactic acid bacteria are gaining global attention, especially due to their role as a probiotic. They are increasingly being used as a flavoring agent and food preservative. Besides their role in food processing, lactic acid bacteria also have a significant role in degrading insecticide residues in the environment. This review paper highlights the importance of lactic acid bacteria in degrading insecticide residues of various types, such as organochlorines, organophosphorus, synthetic pyrethroids, neonicotinoids, and diamides. The paper discusses the mechanisms employed by lactic acid bacteria to degrade these insecticides, as well as their potential applications in bioremediation. The key enzymes produced by lactic acid bacteria, such as phosphatase and esterase, play a vital role in breaking down insecticide molecules. Furthermore, the paper discusses the challenges and future directions in this field. However, more research is needed to optimize the utilization of lactic acid bacteria in insecticide residue degradation and to develop practical strategies for their implementation in real-world scenarios.
Collapse
Affiliation(s)
- K Kiruthika
- Department of Agricultural Entomology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - A Suganthi
- Department of Agricultural Entomology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India.
| | | | - R Anandham
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - P Renukadevi
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - M Murugan
- Department of Agricultural Entomology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - Bimal Kumar Sahoo
- Department of Agricultural Entomology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - Mohammad Ikram
- Department of Agricultural Entomology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - P G Kavitha
- Department of Nematology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| | - M Jayakanthan
- Department of Bioinformatics, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
39
|
Ibrahim M, Bednarczyk M, Stadnicka K, Grochowska E. Inter- and Transgenerational Effects of In Ovo Stimulation with Bioactive Compounds on Cecal Tonsils and Cecal Mucosa Transcriptomes in a Chicken Model. Int J Mol Sci 2025; 26:1174. [PMID: 39940944 PMCID: PMC11817890 DOI: 10.3390/ijms26031174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Exploring how early-life nutritional interventions may impact future generations, this study examines the inter- and transgenerational effects of in ovo injection of bioactive compounds on gene expression in the cecal tonsils and cecal mucosa using a chicken model. Synbiotic PoultryStar® (Biomin) and choline were injected in ovo on the 12th day of egg incubation. Three experimental groups were established in the generation F1: (1) a control group (C) receiving 0.9% physiological saline (NaCl), (2) a synbiotic group (SYN) receiving 2 mg/embryo, and (3) a combined synbiotic and choline group (SYNCH) receiving 2 mg synbiotic and 0.25 mg choline per embryo. For the generations F2 and F3, the SYN and SYNCH groups were each divided into two subgroups: (A) those injected solely in F1 (SYNs and SYNCHs) and (B) those injected in each generation (SYNr and SYNCHr). At 21 weeks posthatching, cecal tonsil and cecal mucosa samples were collected from F1, F2, and F3 birds for transcriptomic analysis. Gene expression profiling revealed distinct intergenerational and transgenerational patterns in both tissues. In cecal tonsils, a significant transgenerational impact on gene expression was noted in the generation F3, following a drop in F2. In contrast, cecal mucosa showed more gene expression changes in F2, indicating intergenerational effects. While some effects carried into F3, they were less pronounced, except in the SYNs group, which experienced an increase compared to F2. The study highlights that transgenerational effects of epigenetic modifications are dynamic and unpredictable, with effects potentially re-emerging in later generations under certain conditions or fading or intensifying over time. This study provides valuable insights into how epigenetic nutritional stimulation during embryonic development may regulate processes in the cecal tonsils and cecal mucosa across multiple generations. Our findings provide evidence supporting the phenomenon of epigenetic dynamics in a chicken model.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
- PBS Doctoral School, Bydgoszcz University of Science and Technology, Aleje prof. S. Kaliskiego 7, 85-796 Bydgoszcz, Poland
| | - Marek Bednarczyk
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| | - Ewa Grochowska
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| |
Collapse
|
40
|
Lv Y, Chu J, Zhang X, Li X, Yin A, on behalf of The Industrial Microbiology Laboratory. Synbiotics effects of d-tagatose and Lactobacillus rhamnosus GG on the inflammation and oxidative stress reaction of Gallus gallus based on the genus of cecal bacteria and their metabolites. PLoS One 2025; 20:e0317825. [PMID: 39869614 PMCID: PMC11771945 DOI: 10.1371/journal.pone.0317825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUNDS Abuse of feed supplement can cause oxidative stress and inflammatory responses in Gallus gallus. Synbiotics are composed of prebiotics and probiotics and it possess huge application potentials in the treatment of animal diseases. METHODS This study examined the effect of d-tagatose on the probiotic properties of L. rhamnosus GG, L. paracasei, and S. lactis so as to screen the best synbiotic combinations. Treat Gallus gallus exhibiting oxidative stress and immune response caused by aflatoxin b1 with optimal synbiotics for 14 days, detect the changes of inflammatory markers and oxidative stress markers of Gallus gallus using qRT-PCR, and identified the intestinal bacteria genera and their metabolites in the cecum of Gallus gallus using gut microbiota and metabolomics analysis. RESULTS AND CONCLUSION The results indicated that oxidative stress and immune response factor expressions quantity in Gallus gallus decreased significantly after 14 days of treatment, compared with model group, the low-dose treatment group's SOD1, SOD3, GPX1, GPX2, GSR, H6DP, and HO-1 genes in liver were downregulated by 36.03%, 40.01%, 45.86%, 40.79%, 37.68%, 25.04%, and 29.89%, the IL-1, IL-2, IL-4, IL-6, IgA, IgM, and IgG genes in blood and spleen were downregulated by 26.59%, 34.19%, 21.19%, 28.18%, 35.93%, 12.67%, 21.81 and 35.93%, 22.85%, 21.19%, 28.78%, 35.93%, 15.36%, 29.73%. The intestinal bacteria genera and metabolomics analysis results indicated that the abundance of beneficial bacteria genus was up-regulated, and the proportion of pathogenic bacteria genera decreased. The amount of beneficial metabolites associated with antioxidant and anti-inflammatory effects was upregulated. The synbiotic composed of d-tagatose and L. rhamnosus GG can treat oxidative stress and immune response by altering the structure of intestinal bacteria genera and the production of metabolites.
Collapse
Affiliation(s)
- Yuanqiang Lv
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Jie Chu
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Xiaoxiao Zhang
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Xuan Li
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Aijiao Yin
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | | |
Collapse
|
41
|
Tain YL, Hsu CN. Does maternal consumption of nutritive and non-nutritive sweeteners result in offspring hypertension? Front Nutr 2025; 12:1464269. [PMID: 39911806 PMCID: PMC11794092 DOI: 10.3389/fnut.2025.1464269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025] Open
Abstract
The consumption of nutritive and non-nutritive sweeteners (NNS) has increased significantly in recent decades. The nutritional status of pregnant women plays a crucial role in determining the likelihood of their offspring developing hypertension in adulthood. While NNSs provide a sweet taste without adding to sugar intake, emerging evidence suggests that maternal consumption of not only nutritive sweeteners (such as fructose) but also NNS may lead to adverse outcomes in offspring, including hypertension. This review provides an overview of the latest research connecting maternal intake of sweeteners to the long-term risk of hypertension in offspring. We examine proposed mechanisms underlying the programming of offspring hypertension by sweeteners, encompassing oxidative stress, dysregulated nutrient sensing signals, abnormal renin-angiotensin system, transcriptome changes, and dysbiotic gut microbiota. Additionally, we outline preventive strategies that can help alleviate offspring hypertension programmed by maternal diets high in sweeteners. Recent advancements in understanding the mechanisms through which maternal consumption of nutritive and non-nutritive sweeteners contributes to offspring hypertension offer promise for addressing this widespread health concern at its developmental roots. Nonetheless, further research is needed to educate the public about the safety of sweetener consumption during pregnancy and lactation.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
42
|
Jang JH, Chang YB, Kim SM, Han K, Sim WS, Hong KB, Suh HJ, Han SH. Impact of the probiotic Bacillus coagulans on loperamide-induced delayed bowel movement in Sprague-Dawley rats. Food Funct 2025; 16:720-730. [PMID: 39745362 DOI: 10.1039/d4fo04237a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
This study investigated the effects of Bacillus coagulans on alleviating loperamide-induced constipation. To evaluate the efficacy of B. coagulans in Sprague-Dawley (SD) rats, fecal parameters, the intestinal transit rate, and changes in intestinal mucosal cells were measured through histological analysis. Additionally, serotonin levels, water absorption, tight junction-related gene expression, and the cecal short-chain fatty acid (SCFA) content were analyzed. The administration of B. coagulans significantly altered the fecal weight and moisture content and improved gastrointestinal transit in rats with loperamide-induced constipation. Furthermore, B. coagulans supplementation restored the thickness of both muscular and mucosal layers that had been reduced by loperamide and significantly increased the area of intestinal cells, including Cajal and crypt cells. B. coagulans administration upregulated the expression levels of tryptophan hydroxylase and aquaporin genes, which were downregulated by loperamide. As the dose of B. coagulans increased, there was a corresponding upregulation in the expression of tight junction-related genes, including occludin (OCLN), zonula occludens 1 (ZO-1), and claudin 1 (CLDN1). Additionally, the levels of c-kit, AQP 3, and OCLN proteins, which were elevated by loperamide treatment, were reduced with higher concentrations of B. coagulans. Loperamide decreased the acetic acid content; however, high doses of B. coagulans increased it, leading to a significant increase in the total cecal SCFA content. Thus, B. coagulans shows potential as a probiotic for improving constipation.
Collapse
Affiliation(s)
- Joo Hyun Jang
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Yeok Boo Chang
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sang Min Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Kisoo Han
- Neo Cremar Co., Ltd., Seoul 06142, Republic of Korea
| | - Wan-Sup Sim
- Neo Cremar Co., Ltd., Seoul 06142, Republic of Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sung Hee Han
- Institute of Human Behavior & Genetics, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
43
|
Sarita B, Samadhan D, Hassan MZ, Kovaleva EG. A comprehensive review of probiotics and human health-current prospective and applications. Front Microbiol 2025; 15:1487641. [PMID: 39834364 PMCID: PMC11743475 DOI: 10.3389/fmicb.2024.1487641] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
The beneficial properties of probiotics have always been a point of interest. Probiotics play a major role in maintaining the health of Gastrointestinal Tract (GIT), a healthy digestive system is responsible for modulating all other functions of the body. The effectiveness of probiotics can be enhanced by formulating them with prebiotics the formulation thus formed is referred to as synbiotics. It not only improves the viability and stability of probiotic cells, but also inhibits the growth of pathogenic strains. Lactobacillus and Bifidobacterium spp. are most commonly used as probiotics. The other microbial spp. that can be used as probiotics are Bacillus, Streptococcus, Enterococcus, and Saccharomyces. Probiotics can be used for the treatment of diabetes, obesity, inflammatory, cardiovascular, respiratory, Central nervous system disease (CNS) and digestive disorders. It is also essential to encapsulate live microorganisms that promote intestinal health. Encapsulation of probiotics safeguards them against risks during production, storage, and gastrointestinal transit. Heat, pressure, and oxidation eradicate probiotics and their protective qualities. Encapsulation of probiotics prolongs their viability, facilitates regulated release, reduces processing losses, and enables application in functional food products. Probiotics as microspheres produced through spray drying or coacervation. This technique regulates the release of gut probiotics and provides stress resistance. Natural encapsulating materials including sodium alginate, calcium chloride, gel beads and polysaccharide promoting safeguards in probiotics during the digestive process. However, several methods including, spray drying where liquid is atomized within a heated air chamber to evaporate moisture and produce dry particles that improves the efficacy and stability of probiotics. Additionally, encapsulating probiotics with prebiotics or vitamins enhance their efficacy. Probiotics enhance immune system efficacy by augmenting the generation of antibodies and immunological cells. It combats illnesses and enhances immunity. Recent studies indicate that probiotics may assist in the regulation of weight and blood glucose levels and influence metabolism and insulin sensitivity. Emerging research indicates that the "gut-brain axis" connects mental and gastrointestinal health. Probiotics may alleviate anxiety and depression via influencing neurotransmitter synthesis and inflammation. Investigations are underway about the dermatological advantages of probiotics that forecasting the onsite delivery of probiotics, encapsulation is an effective technique and requires more consideration from researchers. This review focuses on the applications of probiotics, prebiotics and synbiotics in the prevention and treatment of human health.
Collapse
Affiliation(s)
- Bhutada Sarita
- Department of Microbiology, Sanjivani Arts, Commerce and Science College, Kopargaon, India
| | - Dahikar Samadhan
- Department of Microbiology, Sanjivani Arts, Commerce and Science College, Kopargaon, India
| | - Md Zakir Hassan
- Department of Technologies for Organic Synthesis, Institute of Chemical Technology, Ural Federal University named after the First President of Russia B. N. Yeltsin, Yekaterinburg, Russia
- Bangladesh Livestock Research Institute, Savar, Bangladesh
| | - Elena G. Kovaleva
- Department of Technologies for Organic Synthesis, Institute of Chemical Technology, Ural Federal University named after the First President of Russia B. N. Yeltsin, Yekaterinburg, Russia
| |
Collapse
|
44
|
Azhar Ud Din M, Lin Y, Lyu C, Yi C, Fang A, Mao F. Advancing therapeutic strategies for graft-versus-host disease by targeting gut microbiome dynamics in allogeneic hematopoietic stem cell transplantation: current evidence and future directions. Mol Med 2025; 31:2. [PMID: 39754054 PMCID: PMC11699782 DOI: 10.1186/s10020-024-01060-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a highly effective therapy for malignant blood illnesses that pose a high risk, as well as diseases that are at risk due to other variables, such as genetics. However, the prevalence of graft-versus-host disease (GVHD) has impeded its widespread use. Ensuring the stability of microbial varieties and associated metabolites is crucial for supporting metabolic processes, preventing pathogen intrusion, and modulating the immune system. Consequently, it significantly affects the overall well-being and susceptibility of the host to disease. Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) may experience a disruption in the balance between the immune system and gut bacteria when treated with medicines and foreign cells. This can lead to secondary intestinal inflammation and GVHD. Thus, GM is both a reliable indicator of post-transplant mortality and a means of enhancing GVHD prevention and treatment after allo-HSCT. This can be achieved through various strategies, including nutritional support, probiotics, selective use of antibiotics, and fecal microbiota transplantation (FMT) to target gut microbes. This review examines research advancements and the practical use of intestinal bacteria in GVHD following allo-HSCT. These findings may offer novel insights into the prevention and treatment of GVHD after allo-HSCT.
Collapse
Affiliation(s)
- Muhammad Azhar Ud Din
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yan Lin
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212399, Jiangsu, People's Republic of China
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical College Shangqiu, Shangqiu, 476100, Henan, People's Republic of China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, 212028, Jiangsu, People's Republic of China
| | - Anning Fang
- Basic Medical School, Anhui Medical College, 632 Furong Road, Economic and Technological Development Zone, Hefei, 230061, Anhui, People's Republic of China.
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China.
- Institute of Hematology, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
45
|
De A, Halder S, Madan A, Raja UK, Guru Prasad P, Dhoot D, Jani G, Sharma T. A Comparative, Multicenter, Prospective, Randomized Study to Evaluate the Efficacy, Safety, and Delay of Relapse of Ceramide-Based Post-biotic Moisturizer Versus Paraffin-Based Moisturizer in Mild to Moderate Atopic Dermatitis. Cureus 2025; 17:e76762. [PMID: 39897204 PMCID: PMC11785538 DOI: 10.7759/cureus.76762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic relapsing inflammatory disease that impacts the quality of life of affected individuals as well as their families. Its pathogenesis involves impaired skin barrier function and immune dysregulation. Moisturizers are used in AD management as they help in repairing the skin barrier. OBJECTIVE The objective of this study was to compare the efficacy, safety, and prevention of relapse with ceramide-based post-biotic moisturizer against paraffin-based moisturizer in mild to moderate AD. METHOD A total of 160 AD patients were grouped into two: Group I received desonide cream 0.05% and ceramide-based post-biotic moisturizer and Group II received desonide cream 0.05% and paraffin-based moisturizer. Both groups were given treatment for four weeks. Patients who achieved complete resolution entered the maintenance phase for a maximum duration of three months. They were followed up every two weeks telephonically or as and when the patient experienced a relapse. RESULTS All 160 patients were completely cured and entered the maintenance phase at week 4. A total of 96/160 (71.25%) patients relapsed, with 44 relapses in Group I (55%) and 52 (65%) in Group II (p=0.25). However, in terms of mean relapse time, Group I had a 72.52±15.01 day remission period, whereas Group II had a 47.44±21.49 day remission period (p=0.0001). Moreover, Group I showed a statistically significantly prolonged estimated median time to relapse compared with Group II (median: 85 days versus 71 days, p=0.05). Both moisturizers were tolerated very well. CONCLUSION Although both moisturizers were effective in resolving symptoms in the treatment phase, the ceramide-based post-biotic moisturizer was more effective and statistically significant in extending the remission period against the paraffin-based moisturizer in patients with mild to moderate AD.
Collapse
Affiliation(s)
- Abhishek De
- Dermatology, Wizderm Speciality Skin and Hair Clinic, Kolkata, IND
| | - Saswati Halder
- Dermatology, Calcutta School of Tropical Medicine, Kolkata, IND
| | - Amit Madan
- Dermatology, Ajanta Research Centre, Lucknow, IND
| | | | | | - Dhiraj Dhoot
- Global Medical Affairs, Glenmark Pharmaceuticals Ltd., Mumbai, IND
| | - Gaurang Jani
- Dermatology, Glenmark Pharmaceuticals Ltd., Mumbai, IND
| | - Tripti Sharma
- Global Medical Affairs, Glenmark Pharmaceuticals Ltd., Mumbai, IND
| |
Collapse
|
46
|
Asad A, Kirk M, Zhu S, Dong X, Gao M. Effects of Prebiotics and Probiotics on Symptoms of Depression and Anxiety in Clinically Diagnosed Samples: Systematic Review and Meta-analysis of Randomized Controlled Trials. Nutr Rev 2024:nuae177. [PMID: 39731509 DOI: 10.1093/nutrit/nuae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024] Open
Abstract
CONTEXT The use of prebiotics and probiotics as a treatment for psychiatric conditions has gained interest due to their potential to modulate the gut-brain axis. This review aims to assess the effectiveness of these interventions in reducing symptoms of depression and anxiety in psychiatric populations. OBJECTIVE The aim was to comprehensively review and appraise the effectiveness of prebiotic, probiotic, and synbiotic interventions in reducing clinical depression and anxiety symptoms. DATA SOURCES Systematic searches were conducted across Embase, Medline, PsycINFO, CINAHL, Cochrane Library, and Science Citation Index from database inception to May 22, 2023. DATA EXTRACTION Randomized controlled trials investigating prebiotic, probiotic, or synbiotic interventions for treating clinical depression or anxiety symptoms in clinical samples were included. Data were extracted on study characteristics, intervention details, and outcome measures. The Cochrane Collaboration Tool was used to assess the risk of bias. DATA ANALYSIS The standardized mean difference (SMD) was calculated using Hedge's g as the metric of effect size. A random-effects model was applied to estimate pooled effect sizes with 95% CIs. Subgroup analyses were performed based on study characteristics, methodological factors, and intervention types. Sensitivity analyses excluded studies with a high risk of bias. RESULTS Twenty-three RCTs involving 1401 patients met the inclusion criteria, with 20 trials providing sufficient data for meta-analysis. Of these, 18 trials investigated probiotics for depression, 9 trials assessed probiotics for anxiety, and 3 trials examined prebiotics for depression. Probiotics demonstrated a significant reduction in depression symptoms (SMD: -0.96; 95% CI: -1.31, -0.61) and a moderate reduction in anxiety symptoms (SMD: -0.59; 95% CI: -0.98, -0.19). Prebiotics did not show a significant effect on depression (SMD: -0.28; 95% CI: -0.61, 0.04). High heterogeneity was observed across studies, and subgroup analyses indicated that study duration and probiotic formulations contributed to the variation in effect sizes. CONCLUSION Probiotics showed substantial reductions in depression symptoms and moderate reductions in anxiety symptoms. Prebiotics showed a nonsignificant trend toward reducing depression. An adjunctive mental health treatment approach that diagnoses, monitors, and treats the gut microbiome alongside traditional pharmacological treatment holds promise for clinical practice. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023424136.
Collapse
Affiliation(s)
- Afrida Asad
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, United Kingdom
| | - Megan Kirk
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
- NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| | - Sufen Zhu
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Xue Dong
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Min Gao
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
- NIHR Oxford Health Biomedical Research Centre, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| |
Collapse
|
47
|
Djenane D, Khaled BM, Ben Miri Y, Metahri MS, Montañés L, Aider M, Ariño A. Improved Functionality, Quality, and Shelf Life of Merguez-Type Camel Sausage Fortified with Spirulina as a Natural Ingredient. Foods 2024; 14:59. [PMID: 39796348 PMCID: PMC11719629 DOI: 10.3390/foods14010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The objective of the present work was to examine the effect of incorporating spirulina powder (SP) in merguez-type sausages made exclusively with camel meat, as well as to evaluate its physicochemical, microbiological, and sensory quality attributes and its prebiotic potential. The final purpose was to offer an innovative meat product to increase camel meat consumption. Several innovative fresh sausage formulations were developed using SP (00, 100, 250, and 500 mg/kg) and stored under vacuum conditions with refrigeration at 1 ± 1 °C for 35 days. A control group of camel sausage without SP was also stored overwrapped (OW) under aerobic conditions, to serve as the negative control. The addition of SP to the vacuum-packed camel sausages extended their shelf life by 20 to 35 days compared to the control group, which was completely spoiled by the fifth day of storage. These results were more pronounced the higher the percentage of SP incorporated into the camel sausage formulation, as indicated by the following parameters: 2-thiobarbituric acid-reactive substances TBARS (1.46 vs. 2.89 mg MDA/kg), CIE a* (14.65 vs. 10.12), total volatile basic nitrogen TVB-N (13.02 vs. 15.09 mg/kg), total psychrotrophic bacteria TPB (5.71 vs. 6.34 log CFU/g), and overall acceptability score (3.17 vs. 2.5). The study of prebiotic potential suggested that the addition of SP to camel sausages promoted the growth of probiotic strains, which in turn were able to inhibit the growth of pathogenic microorganisms such as S. aureus and E. coli O157:H7. In conclusion, this study highlighted how SP, as a clean label ingredient, based on its rich composition and its antioxidant, antibacterial, and prebiotic effects, may represent a source of beneficial substances for human health and offer an alternative approach to producing a new traditional merguez-type sausage with improved acceptance.
Collapse
Affiliation(s)
- Djamel Djenane
- Meat Quality and Meat Safety Laboratory, University Mouloud Mammeri, Tizi Ouzou 15000, Algeria;
| | - Boumediène Méghit Khaled
- Laboratoire de Nutrition, Pathologie, Agro-Biotechnologie et Santé (Lab-NuPABS), Department of Biology, Faculty of Natural Sciences and Life, Djillali Liabès University, Sidi Bel Abbès 22005, Algeria;
| | - Yamina Ben Miri
- Meat Quality and Meat Safety Laboratory, University Mouloud Mammeri, Tizi Ouzou 15000, Algeria;
- Department of Biochemistry and Microbiology, Faculty of Sciences, Mohamed Boudiaf University, M’sila 28000, Algeria
| | - Mohammed Said Metahri
- Faculty of Biological and Agricultural Sciences, Mouloud Mammeri University of Tizi-Ouzou, Tizi Ouzou 15000, Algeria;
| | - Luis Montañés
- Laboratorios Valero Analítica S.L., 50011 Zaragoza, Spain;
| | - Mohammed Aider
- Department of Soil Sciences and Agri-Food Engineering, Université Laval, Quebec City, QC G1V 0A6, Canada;
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Agustín Ariño
- Facultad de Veterinaria, Instituto Agroalimentario de Aragón-IA2, Universidad de Zaragoza-CITA, 50013 Zaragoza, Spain;
| |
Collapse
|
48
|
Sepehr A, Miri ST, Aghamohammad S, Rahimirad N, Milani M, Pourshafie MR, Rohani M. Health benefits, antimicrobial activities, and potential applications of probiotics: A review. Medicine (Baltimore) 2024; 103:e32412. [PMID: 39969286 PMCID: PMC11688011 DOI: 10.1097/md.0000000000032412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 12/02/2022] [Indexed: 02/20/2025] Open
Abstract
Gut microbiota and its metabolic activities can influence the physiology and pathology of the human body. It is well established that alterations in the balance of living microbiota can contribute to various health problems, such as inflammatory bowel disease and autoimmune disorders. Probiotics administered in sufficient quantities as functional food ingredients provide health benefits to hosts. They help to maintain the stability and composition of the gut microbiota and provide resistance to infection by pathogens. The most important probiotic bacteria are Lactobacillus spp. and Bifidobacteria spp., which protect the intestine through various mechanisms such as the production of organic acids and bacteriocins. Scientific and clinical research has demonstrated that probiotics play a role in modulating immune response and preventing cancer and chronic inflammatory diseases, especially in the gastrointestinal tract. This article summarizes the potential health benefits, antimicrobial activities, and purposes for which probiotics can be used as functional foods to improve human health.
Collapse
Affiliation(s)
- Amin Sepehr
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Tina Miri
- Department of Microbiology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | | | - Nazanin Rahimirad
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahnaz Milani
- Department of Microbiology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | | | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
49
|
Tain YL, Hsu CN. Kidney Programming and Hypertension: Linking Prenatal Development to Adulthood. Int J Mol Sci 2024; 25:13610. [PMID: 39769369 PMCID: PMC11677590 DOI: 10.3390/ijms252413610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
The complex relationship between kidney disease and hypertension represents a critical area of research, yet less attention has been devoted to exploring how this connection develops early in life. Various environmental factors during pregnancy and lactation can significantly impact kidney development, potentially leading to kidney programming that results in alterations in both structure and function. This early programming can contribute to adverse long-term kidney outcomes, such as hypertension. In the context of kidney programming, the molecular pathways involved in hypertension are intricate and include epigenetic modifications, oxidative stress, impaired nitric oxide pathway, inappropriate renin-angiotensin system (RAS) activation, disrupted nutrient sensing, gut microbiota dysbiosis, and altered sodium transport. This review examines each of these mechanisms and highlights reprogramming interventions proposed in preclinical studies to prevent hypertension related to kidney programming. Given that reprogramming strategies differ considerably from conventional treatments for hypertension in kidney disease, it is essential to shift focus toward understanding the processes of kidney programming and its role in the development of programmed hypertension.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
50
|
Todorov SD, Tagg J, Algburi A, Tiwari SK, Popov I, Weeks R, Mitrokhin OV, Kudryashov IA, Kraskevich DA, Chikindas ML. The Hygienic Significance of Microbiota and Probiotics for Human Wellbeing. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10419-9. [PMID: 39688648 DOI: 10.1007/s12602-024-10419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 12/18/2024]
Abstract
The human body can be viewed as a combination of ecological niches inhabited by trillions of bacteria, viruses, fungi, and parasites, all united by the microbiota concept. Human health largely depends on the nature of these relationships and how they are built and maintained. However, personal hygiene practices have historically been focused on the wholesale elimination of pathogens and "hygiene-challenging microorganisms" without considering the collateral damage to beneficial and commensal species. The microbiota can vary significantly in terms of the qualitative and quantitative composition both between different people and within one person during life, and the influence of various environmental factors, including age, nutrition, bad habits, genetic factors, physical activity, medication, and hygienic practices, facilitates these changes. Disturbance of the microbiota is a predisposing factor for the development of diseases and also greatly influences the course and severity of potential complications. Therefore, studying the composition of the microbiota of the different body systems and its appropriate correction is an urgent problem in the modern world. The application of personal hygiene products or probiotics must not compromise health through disruption of the healthy microbiota. Where changes in the composition or metabolic functions of the microbiome may occur, they must be carefully evaluated to ensure that essential biological functions are unaffected. As such, the purpose of this review is to consider the microbiota of each of the "ecological niches" of the human body and highlight the importance of the microbiota in maintaining a healthy body as well as the possibility of its modulation through the use of probiotics for the prevention and treatment of certain human diseases.
Collapse
Affiliation(s)
- Svetoslav D Todorov
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos E Nutrição Experimental, Food Research Center, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil.
- Instituto Politécnico de Viana Do Castelo, 4900-347, Viana Do Castelo, Portugal.
| | - John Tagg
- Blis Technologies, South Dunedin, 9012, New Zealand
| | - Ammar Algburi
- Department of Microbiology, College of Veterinary Medicine, University of Diyala, Baqubah, Iraq
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Igor Popov
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Olimpijskij Ave., 1, Federal Territory Sirius, Sirius, 354340, Russia
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Villafloraweg, 1, 5928 SZ, Venlo, The Netherlands
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Oleg V Mitrokhin
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Ilya A Kudryashov
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Denis A Kraskevich
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia.
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|