1
|
Nama ASA, Sandeepa GM, Buddolla V, Mastan A. Advances in understanding therapeutic mechanisms of probiotics in cancer management, with special emphasis on breast cancer: A comprehensive review. Eur J Pharmacol 2025; 995:177410. [PMID: 39986595 DOI: 10.1016/j.ejphar.2025.177410] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
The increasing global prevalence of cancer, particularly breast cancer, necessitates the development of innovative therapeutic strategies. Probiotics, proficient in promoting gut health, have emerged as promising candidates for cancer treatment due to their immunomodulatory and potential anticancer properties. This review focuses on the therapeutic mechanisms of probiotics in breast cancer, examining their anticancer efficacy through metabolic, immune, and molecular mechanisms. Probiotics enhance cancer therapies, minimize side effects, and offer new adjuvant approaches in oncology. Recent advancements discussed in the review include the utilization of probiotics as oncolytic gene expression systems and drug delivery vectors, as well as personalized probiotic interventions aimed at optimizing cancer therapy. Clinical studies are critically evaluated, highlighting both the outcomes and limitations of probiotic use in cancer patients, particularly those suffering from breast cancer. Additionally, the review explores factors influencing anticancer effects of probiotics, focusing on their role in modulating the tumor microenvironment. Challenges in translating preclinical findings to clinical practice are discussed, along with future research directions, focusing on the relationship between probiotics, the microbiome, and cancer treatment. Ultimately, this review advocates for further investigation into the therapeutic potential of probiotics in breast cancer, aiming to harness their benefits in oncology.
Collapse
Affiliation(s)
- A S Angel Nama
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India
| | - G Mary Sandeepa
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India.
| | - Viswanath Buddolla
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India
| | - Anthati Mastan
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India.
| |
Collapse
|
2
|
Cebi M, Yilmaz Y. Epithelial barrier hypothesis in the context of nutrition, microbial dysbiosis, and immune dysregulation in metabolic dysfunction-associated steatotic liver. Front Immunol 2025; 16:1575770. [PMID: 40438102 PMCID: PMC12116361 DOI: 10.3389/fimmu.2025.1575770] [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: 02/12/2025] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
In recent years, the prevalence of chronic liver diseases, particularly Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), has increased significantly. This upward trend is largely associated with lifestyle-related factors such as unhealthy dietary habits, physical inactivity, and various environmental influences. Among the key elements contributing to the pathogenesis of MASLD, the integrity of the intestinal epithelial barrier emerges as a critical determinant, given its central role in maintaining immune homeostasis along the gut-liver axis. Disruption of this barrier, often driven by excessive consumption of saturated fats and refined carbohydrates in combination with low dietary fiber intake, can lead to microbial dysbiosis. This imbalance in the gut microbiota triggers immune dysregulation and promotes systemic inflammation, thereby exacerbating hepatic injury. This review discusses the contribution of epithelial barrier dysfunction to the development and progression of MASLD, with a particular focus on how increased intestinal permeability may initiate and sustain chronic liver inflammation. Additionally, the influence of dietary and environmental factors on epithelial integrity, immune responses, and the inflammatory cascade is addressed. A better understanding of the complex interplay between gut barrier impairment, immune modulation, and liver pathology may offer valuable insights into MASLD pathophysiology and contribute to the development of more targeted therapeutic strategies.
Collapse
Affiliation(s)
- Merve Cebi
- Department of Medical Biology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
- The Global NASH Council, Washington, DC, United States
| |
Collapse
|
3
|
Chaves LS, Oliveira ACP, Oliveira AP, Lopes ALF, Araujo AKS, Pacheco G, Silva KC, Martins FEC, Gomes IAB, Ramos SVS, Viana HTMC, Batista AVF, Oliveira BC, Nicolau LAD, Ribeiro FOS, Castro AV, de Araujo-Nobre AR, Silva DA, Cordeiro LMC, Góis MB, Medeiros JVR. Cashew gum fractions protect intestinal mucosa against shiga toxin-producing Escherichia coli infection: Characterization and insights into microbiota modulation. Int J Biol Macromol 2025; 311:143916. [PMID: 40324507 DOI: 10.1016/j.ijbiomac.2025.143916] [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: 02/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Diarrheal diseases remain a major public health concern, particularly in regions with poor sanitation. Polysaccharides extracted from natural gums have been investigated as functional agents for intestinal health, and their fractionation enables the production of oligosaccharides with potential prebiotic activity. This study aimed to produce cashew gum (CG) fractions through Smith degradation (CGD48) and partial hydrolysis (CGD24) and to evaluate their ability to modulate and protect the intestinal microbiota. Balb/c mice were administered CG (1200 mg/kg), CGD24 (800 mg/kg), or CGD48 (800 mg/kg) for 10 and 26 days, followed by infection with Shiga toxin-producing Escherichia coli (STEC) (5 × 1010 CFU/mL) for three days. Characterization assays confirmed the fragmentation of CG. Both CGD24 and CGD48 promoted the growth of beneficial bacteria with and without infection and reduced STEC colonization. Furthermore, they preserved mucin levels in the cecum and large intestine and maintained baseline levels of superoxide dismutase (SOD), suggesting protection of the intestinal mucosa. These findings indicate that CG fractions exhibit microbiota-modulating and protective effects against STEC, highlighting their therapeutic potential and the need for further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Letícia S Chaves
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Antonio C P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - André L F Lopes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Andreza K S Araujo
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Gabriella Pacheco
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Katriane C Silva
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Francisco E C Martins
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Isaac A B Gomes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Sabrine V S Ramos
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Hémilly T M C Viana
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana V F Batista
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Beatriz C Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucas A D Nicolau
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Fábio O S Ribeiro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Auricélia V Castro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Alyne Rodrigues de Araujo-Nobre
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Durcilene A Silva
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo B Góis
- Post-Graduation Program in Biosciences and Health, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Jand V R Medeiros
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil; Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil.
| |
Collapse
|
4
|
Mafe AN, Nkene IH, Ali ABM, Edo GI, Akpoghelie PO, Yousif E, Isoje EF, Igbuku UA, Ismael SA, Essaghah AEA, Ahmed DS, Umar H, Alamiery AA. Smart Probiotic Solutions for Mycotoxin Mitigation: Innovations in Food Safety and Sustainable Agriculture. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10569-4. [PMID: 40312537 DOI: 10.1007/s12602-025-10569-4] [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: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Mycotoxin contamination poses severe risks to food safety and agricultural sustainability. Probiotic-based interventions offer a promising strategy for mitigating these toxic compounds through adsorption, biodegradation, and gut microbiota modulation. This review examines the mechanisms by which specific probiotic strains inhibit mycotoxin biosynthesis, degrade existing toxins, and enhance host detoxification pathways. Emphasis is placed on strain-specific interactions, genetic and metabolic adaptations, and advancements in formulation technologies that improve probiotic efficacy in food matrices. Also, the review explores smart delivery systems, such as encapsulation techniques and biofilm applications, to enhance probiotic stability and functionality. Issues related to regulatory approval, strain viability, and large-scale implementation are also discussed. By integrating molecular insights, applied case studies, and innovative probiotic-based solutions, this review provides a roadmap for advancing safe and sustainable strategies to combat mycotoxin contamination in food and agricultural systems.
Collapse
Affiliation(s)
- Alice Njolke Mafe
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Jalingo, Nigeria
| | - Istifanus Haruna Nkene
- Department of Microbiology, Faculty of Natural and Applied Sciences, Nasarawa State University, Keffi, Nigeria
| | - Ali B M Ali
- Department of Air Conditioning Engineering, College of Engineering, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Great Iruoghene Edo
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria.
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq.
| | - Patrick Othuke Akpoghelie
- Department of Food Science and Technology, Faculty of Science, Delta State University of Science and Technology, Ozoro, Delta State, Nigeria
| | - Emad Yousif
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Endurance Fegor Isoje
- Department of Biochemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Ufuoma Augustina Igbuku
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Shams A Ismael
- Department of Medical Physics, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Arthur Efeoghene Athan Essaghah
- Department of Urban and Regional Planning, Faculty of Environmental Sciences, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Dina S Ahmed
- Department of Chemical Industries, Institute of Technology-Baghdad, Middle Technical University, Baghdad, Iraq
| | - Huzaifa Umar
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus
| | - Ahmed A Alamiery
- AUIQ, Al-Ayen Scientific Research Center, Al-Ayen Iraqi University, Thi Qar, P.O. Box: 64004, An Nasiriyah, Iraq
| |
Collapse
|
5
|
Dibakoane SR, Mhlongo G, Moonsamy G, Wokadala OC, Mnisi CM, Mlambo V. Phenomenological and mechanistic insights into potential dietary nucleotide - probiotic synergies in layer chickens: A review. Poult Sci 2025; 104:105049. [PMID: 40106904 PMCID: PMC11964621 DOI: 10.1016/j.psj.2025.105049] [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: 02/08/2025] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025] Open
Abstract
Despite their growing popularity as alternatives to antibiotic growth promoters (AGPs), the individual effects of nucleotides and probiotics on poultry gut functionality remain poorly understood. In addition, inconsistent outcomes are quite common in studies where these two additives have been used separately to modify gut function and related parameters in birds. These inconsistencies, which have limited the potential of probiotics and nucleotides as AGP replacements, stem from various factors and need to be addressed. Combining probiotics and nucleotides could potentially enhance their effectiveness and lead to more consistent outcomes in layer chickens. Since their mechanisms of action complement each other, some level of synergy is expected when used together. Both additives have been shown to support gut health, boost immune function, and improve performance in chickens when used individually. However, no studies have investigated the possible synergistic effects of nucleotides and probiotics in poultry. This review makes the case for combined use of probiotics and nucleotides in layer chickens by providing phenomenological and mechanistic insights into hypothetical synergistic effects. This paper highlights the need for AGP alternatives and reviews studies on the effects and mechanisms of probiotics and nucleotides in layer chickens when used individually. We then propose potential mechanisms for their synergistic effects on gut health, performance, and egg quality based on logical deductions from observed biological responses. These proposed mechanisms are hypothetical and require experimental validation. Finally, the review explores how this synergy could lead to more consistent outcomes and enhance the feasibility of AGP-free egg production.
Collapse
Affiliation(s)
- Siphosethu R Dibakoane
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Godfrey Mhlongo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Ghaneshree Moonsamy
- Council for Scientific and Industrial Research (CSIR); Future production: Chemicals, Meiring Naude Drive, Pretoria 0081, South Africa
| | - Obiro Cuthbert Wokadala
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Caven Mguvane Mnisi
- Department of Animal Science, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa; Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa
| | - Victor Mlambo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa.
| |
Collapse
|
6
|
Ma W, Wu Y, Sun H, Zhao Y, Huang L. An in vivo study of the ameliorative effect of supplementation with Lacticaseibacillus paracasei Glory LP16 in immunocompromised mice. Sci Rep 2025; 15:14932. [PMID: 40295599 PMCID: PMC12037797 DOI: 10.1038/s41598-025-97430-4] [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: 08/20/2024] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Probiotics play a beneficial role in improving the intestinal microecological balance and improving the health level and state of the host. In this study, cyclophosphamide was used to establish an immunocompromised mouse model. In the experiment, sixty mice were randomly divided into 5 groups: normal control, model control, Glory LP16 low-dose control (2 × 106 CFU/mouse, LP), Glory LP16 medium-dose control (2 × 107 CFU/mouse, MP), and Glory LP16 high-dose control (2 × 108 CFU/mouse, HP). The mice were tested for body weight, immune organ indexes, cellular immunity indexes, humoral immunity indexes, non-specific immunity indexes, colonic histopathological notices, intestinal flora, and short-chain fatty acids. The results showed that compared with the model control, the high-dose control showed an increase in body mass gain, thymus index, spleen index, optical density index, foot-plantar thickness, number of hemolyzed vacuoles, number of antibody accumulation, NK cell activity, carbon profile phagocytosis index, macrophage neutral red phagocytosis rate, macrophage phagocytosis index, the morphology of colon tissue tended to be more like that of the normal tissue, the regulation of the intestinal bacterial flora imbalance, and an increase in short-chain fatty acids of the intestine. It is hypothesized that Lacticaseibacillus paracasei Glory LP16 has the function of enhancing immunity in mice.
Collapse
Affiliation(s)
- Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150030, Heilongjiang, China
| | - Yanan Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150030, Heilongjiang, China
| | - Hang Sun
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150030, Heilongjiang, China
| | - Yiyang Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150030, Heilongjiang, China
| | - Lili Huang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, 150030, Heilongjiang, China.
| |
Collapse
|
7
|
Lu H, Zhang G, Wang K, Liu K, Gao Y, Chen J, Li Y, Yan J. The Role of Lactiplantibacillus plantarum CGMCC9513 in Alleviating Colitis by Synergistic Enhancement of the Intestinal Barrier Through Modulating Gut Microbiota and Activating the Aryl Hydrocarbon Receptor. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10551-0. [PMID: 40301232 DOI: 10.1007/s12602-025-10551-0] [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: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Ulcerative colitis (UC) has become a global health issue. This study evaluated whether administering Lactiplantibacillus plantarum CGMCC9513 to dextran sulfate sodium (DSS)-induced colitis mice could alleviate colitis by modulating gut microbiota imbalance and activating the aryl hydrocarbon receptor (AhR) to enhance intestinal barrier function. The anti-inflammatory effect and AhR activation ability of L. plantarum CGMCC9513 were evaluated with lipopolysaccharide (LPS)-induced cell inflammation model; 25 male BALB/c mice were divided into blank group (CNG), model group (DSS), L. plantarum CGMCC9513-treated group (LPDT), and L. plantarum CGMCC9513 control group (LP). The mice were pre-administered L. plantarum CGMCC9513 for 14 days and continued to receive it during DSS induction. Symptoms during induction, goblet cell count, expression of MUC2 and Occludin proteins, and changes in gut microbiota were observed. Subsequently, the expression of cytokines interleukin-10 (IL-10), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β) and AhR activation status was determined. The study found that L. plantarum CGMCC9513 could alleviate cell inflammation induced by LPS and activate AhR in vitro. For colitis mice, it could reduce colonic mucosal damage and enhance intestinal barrier function. Regarding gut microbiota changes, L. plantarum CGMCC9513 mainly downregulated Bacteroides, Blautia, Escherichia-Shigella, and Lachnospiraceae_ NK4A136_group and upregulated Firmicutes, Lactobacillus. It reduces the risk of bacterial translocation and increases beneficial gut bacteria. L. plantarum CGMCC9513 reduced the expression of pro-inflammatory cytokines TNF-α and IL-1β while increasing the expression of anti-inflammatory cytokine IL-10. Meanwhile, increased expression of AhR and Cytochrome P450 1A1 (CYP1A1) proteins indicated AhR activation by L. plantarum CGMCC9513. In conclusion, L. plantarum CGMCC9513 can synergistically enhance intestinal barrier alleviation in colitis mice by modulating gut microbiota imbalance and activating AhR.
Collapse
Affiliation(s)
- Hongyu Lu
- Medical School of Guangxi University, Nanning, 530004, China
| | - Guoqing Zhang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kaidi Wang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yingrui Gao
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Jinyan Chen
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yixiang Li
- Medical School of Guangxi University, Nanning, 530004, China.
| | - Jianhua Yan
- Medical School of Guangxi University, Nanning, 530004, China.
| |
Collapse
|
8
|
Bertola B, Cotolí-Crespo A, San Onofre N, Soriano JM. The Mystery of Certain Lactobacillus acidophilus Strains in the Treatment of Gastrointestinal Symptoms of COVID-19: A Review. Microorganisms 2025; 13:944. [PMID: 40284780 PMCID: PMC12029412 DOI: 10.3390/microorganisms13040944] [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: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
COVID-19 presents a wide range of symptoms, including gastrointestinal manifestations such as diarrhea, nausea, and abdominal pain. Lactobacillus acidophilus has been proposed as a potential adjunct therapy to alleviate these symptoms due to its probiotic properties, which help restore gut microbiota balance and modulate immune responses. This review systematically analyzed studies assessing the effects of L. acidophilus in COVID-19 patients with gastrointestinal symptoms. The literature search was conducted through PubMed and the WHO COVID-19 database using keywords such as "Lactobacillus acidophilus", "COVID-19", "gastrointestinal symptoms", and "inflammation markers". The search covered studies published until February 2025. Inclusion criteria: observational and clinical trials with L. acidophilus for symptom relief. Exclusion: animal studies and non-ethical approvals. The findings suggest that L. acidophilus supplementation may contribute to faster resolution of diarrhea, improved gut microbiota balance, and reduced inflammatory markers. However, some studies have found no significant impact on hospitalization rates or disease progression. The probiotic's mechanisms of action appear to involve microbiota modulation, intestinal barrier reinforcement, and anti-inflammatory effects rather than direct viral inhibition in COVID-19 after progression. Some L. acidophilus strains show promise, and clinical validation should follow careful preclinical studies (in vitro, cell lines, and animal models), especially in vulnerable populations such as immunocompromised individuals. Understanding the gut-lung axis and its role in immune response regulation, together with the need for a thorough characterization of the specific strains, including biochemical, genomic, and functional properties, before testing in humans, may provide deeper insights into the therapeutic potential of probiotics in viral infections.
Collapse
Affiliation(s)
- Belén Bertola
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Paterna, Spain;
| | - Amparo Cotolí-Crespo
- Faculty of Psychology and Speech Therapy, University of Valencia, 46010 Valencia, Spain;
| | - Nadia San Onofre
- NUTRALiSS Research Group, Faculty of Health Sciences, Universitat Oberta de Catalunya, Rambla del Poblenou 156, 08018 Barcelona, Spain;
| | - Jose M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Paterna, Spain;
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute La Fe, 46026 Valencia, Spain
| |
Collapse
|
9
|
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
|
10
|
Abdul Manan M. Progress in Probiotic Science: Prospects of Functional Probiotic-Based Foods and Beverages. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2025; 2025:5567567. [PMID: 40259922 PMCID: PMC12011469 DOI: 10.1155/ijfo/5567567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
This comprehensive review explores the evolving role of probiotic-based foods and beverages, highlighting their potential as functional and "future foods" that could significantly enhance nutrition, health, and overall well-being. These products are gaining prominence for their benefits in gut health, immune support, and holistic wellness. However, their future success depends on addressing critical safety concerns and navigating administrative complexities. Ensuring that these products "do more good than harm" involves rigorous evaluations of probiotic strains, particularly those sourced from the human gastrointestinal tract. Lactic acid bacteria (LABs) serve as versatile and effective functional starter cultures for the development of probiotic foods and beverages. The review emphasizes the role of LABs as functional starter cultures and the development of precision probiotics in advancing these products. Establishing standardized guidelines and transparent practices is essential, requiring collaboration among regulatory bodies, industry stakeholders, and the scientific community. The review underscores the importance of innovation in developing "friendly bacteria," "super probiotics," precision fermentation, and effective safety assessments. The prospects of functional probiotic-based foods and beverages rely on refining these elements and adapting to emerging scientific advancements. Ultimately, empowering consumers with accurate information, fostering innovation, and maintaining stringent safety standards will shape the future of these products as trusted and beneficial components of a health-conscious society. Probiotic-based foods and beverages, often infused with LABs, a "friendly bacteria," are emerging as "super probiotics" and "future foods" designed to "do more good than harm" for overall health.
Collapse
Affiliation(s)
- Musaalbakri Abdul Manan
- Food Science and Technology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), MARDI Headquarters, Persiaran MARDI-UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
11
|
Sun J, Wang X, He Y, Han M, Li M, Wang S, Chen J, Zhang Q, Yang B. Environmental fate of antibiotic resistance genes in livestock farming. Arch Microbiol 2025; 207:120. [PMID: 40214801 DOI: 10.1007/s00203-025-04320-4] [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: 02/08/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
As emerging environmental pollutants, antibiotic resistance genes (ARGs) are prevalent in livestock farms and their surrounding environments. Although existing studies have focused on ARGs in specific environmental media, comprehensive research on ARGs within farming environments and their adjacent areas remains scarce. This review explores the sources, pollution status, and transmission pathways of ARGs from farms to the surrounding environment. Drawing on the "One Health" concept, it also discusses the potential risks of ARGs transmission from animals to human pathogens and the resulting impact on human health. Our findings suggest that the emergence of ARGs in livestock farming environments primarily results from intrinsic resistance and genetic mutations, while their spread is largely driven by horizontal gene transfer. The distribution of ARGs varies according to the type of resistance genes, seasonal changes, and the medium in which they are present. ARGs are disseminated into the surrounding environment via pathways such as manure application, wastewater discharge, and aerosol diffusion. They may be absorbed by humans, accumulating in the intestinal microbiota and subsequently affecting human health. The spread of ARGs is influenced by the interplay of microbial communities, antibiotics, heavy metals, emerging pollutants, and environmental factors. Additionally, we have outlined three control strategies: reducing the emergence of ARGs at the source, controlling their spread, and minimizing human exposure. This article provides a theoretical framework and scientific guidance for understanding the cross-media migration of microbial resistance in livestock farming environments.
Collapse
Affiliation(s)
- Jiali Sun
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China
| | - Xiaoqi Wang
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China
| | - Yuanjie He
- College of Life Science, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Mengting Han
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China
| | - Mei Li
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China
| | - Siyue Wang
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China
| | - Jia Chen
- Shijiazhuang University, Shijiazhuang, 050035, China
| | - Qiang Zhang
- National Institutes for Food and Drug Control, Beijing, 100050, China.
| | - Baowei Yang
- College of Food Science and Engineering, Northwest A&F University, 22# Xi'nong Road, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
12
|
Liu F, Yu J, Chen Z, Zhang S, Zhang Y, Zhang L, Zhang Y, Li J, Ding L, Wu J. Isolation of Bacillus cereus and its probiotic effect on growth performance, antioxidant capacity, and intestinal barrier protection of broilers. Poult Sci 2025; 104:104944. [PMID: 40014971 PMCID: PMC11910085 DOI: 10.1016/j.psj.2025.104944] [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: 12/17/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025] Open
Abstract
Probiotics are effective for improving poultry health. Probiotic Bacillus cereus strains are widely used to improve animal health by stimulating the immune system. In this study, we obtained a B. cereus 13 (BC13) strain that functions in acid, high-temperature, and bile salt resistance. It also degrades starch, cellulose, and other proteins. To better understand the probiotic effects of BC13, we added the strain to the diet of broilers and observed its effects. We found that BC13 significantly improved the growth performance of broilers. The levels of total antioxidant capacity, superoxide dismutase, and glutathione peroxidase were increased, and the concentration of malondialdehyde was reduced by BC13. Supplementation with BC13 enhanced immune function by increasing the levels of secretory immunoglobulin A (sIgA) in the jejunum mucosa; IgA, IgM, and IgG in the serum; mRNA levels of Zo-1, claudin and occludin of the jejunal mucosa; and increased villus height/crypt depth of the jejunum. Furthermore, BC13 improved the composition of intestinal microbes, especially at the genus level of Akkermansia. The addition of BC13 increased the levels of acetic, butyric, valeric, and propionic acids. These results emphasise the potential of BC13 as a probiotic dietary supplement to improve the antioxidant capacity, intestinal barrier function, and gut microbial composition to enhance body health.
Collapse
Affiliation(s)
- Fei Liu
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Jiang Yu
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China; School of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Zhi Chen
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Shuzhi Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Yue Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Lin Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Yuyu Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China; School of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Jianda Li
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Luogang Ding
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China
| | - Jiaqiang Wu
- Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, PR China; School of Life Sciences, Shandong Normal University, Jinan, 250014, PR China.
| |
Collapse
|
13
|
He Q, Li M, Diao H, Zheng Q, Li M, Zhu Q, Cui W. Association of Dietary Live Microbe Intake With Mortality: Results From the National Health and Nutrition Examination Survey, 1999-2018. J Acad Nutr Diet 2025:S2212-2672(25)00109-1. [PMID: 40147756 DOI: 10.1016/j.jand.2025.03.007] [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: 06/21/2024] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The association between dietary intake of live microbes and mortality remains unclear. OBJECTIVE This study aims to investigate the relationship between dietary live microbial intake and all-cause and cause-specific mortality among adults in the United States. DESIGN This is a cross-sectional study of adults aged 20 years or older who participated in the 1999-2018 National Health and Nutrition Examination Survey. PARTICIPANTS AND SETTING The study utilized data from adults aged 20 years and older with complete dietary and mortality data from the National Health and Nutrition Examination Survey from 1999 to 2018. MAIN OUTCOME MEASURES Deaths from any cause are defined as all-cause mortality. The International Statistical Classification of Diseases, 10th Revision, and the National Center for Health Statistics classifications of heart disease (054-064) and malignant neoplasms (019-043) were used to identify disease-specific causes of death. STATISTICAL ANALYSES PERFORMED Cox proportional hazard regression was utilized to examine the associations between the consumption of dietary live microbes and all-cause and cause-specific mortality. Restricted cubic spline regression modeling was used to assess potential linear associations between dietary live microorganism intake and mortality. In addition, stratified analyses and sensitivity analyses of the association of dietary live microorganism intake with all-cause and cardiovascular deaths were performed to validate the robustness of the results. RESULTS The study included 31 836 participants, of whom 4160 died, including 1109 cardiovascular deaths and 915 cancer deaths. The study found that consuming live microbes from the diet was linked to a lower rate of all-cause and cardiovascular mortality, respectively (hazard ratio 0.80, 95% CI 0.72 to 0.89; P < .001; hazard ratio 0.79, 95% CI 0.65 to 0.95; P = .014). However, there was no significant association observed between microbial intake and cancer mortality (hazard ratio 0.93, 95% CI 0.75 to 1.17; P = .545). Restricted cubic spline demonstrates a linear association between dietary live microorganism intake and all-cause and cardiovascular mortality (P < .001). Furthermore, sensitivity analyses indicated that a high intake of live dietary microorganisms was associated with a lower risk of all-cause mortality and cardiovascular mortality (P < .05). CONCLUSIONS The study found that consuming live microbes through diet was linked to a lower rate of all-cause and cardiovascular mortality but not cancer mortality.
Collapse
Affiliation(s)
- Qingzhen He
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Mingshuo Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Houze Diao
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Qingzhao Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Mingyuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Qing Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, PR China.
| |
Collapse
|
14
|
Jia H, Xie Y, Yi L, Cheng W, Song G, Shi W, Zhu J, Zhao S. Comparative Analysis of Short-Chain Fatty Acids and the Immune Barrier in Cecum of Dahe Pigs and Dahe Black Pigs. Animals (Basel) 2025; 15:920. [PMID: 40218314 PMCID: PMC11987949 DOI: 10.3390/ani15070920] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
The intestinal immune barrier is a developed and complex immune system, and there is a fine synergy between it and the induced immune response. Short-chain fatty acids (SCFAs) are the main metabolites of intestinal microbial fermentation. In the cecum of pigs, SCFAs not only provide energy for the host but also participate in regulating the function of the intestinal immune system. The purpose of this study was to explore the mechanism of SCFAs in the regulation of immune gene expression in porcine cecum. SCFAs content and mRNA expression levels of immune genes in cecum were detected, and Gene Ontology (GO) function annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, Protein-Protein Interaction Networks (PPI) network construction, key gene identification, and correlation analysis were performed. The results showed that the content of SCFAs in the cecum of Dahe black pigs (DHB) was lower than that of Dahe pigs (DH). There were significant differences in mRNA expression of some immune genes between the two groups. GO functional annotation found terms related to cytokine activity and protein heterodimerization activity; the KEGG pathway was enriched in several pathways related to intestinal immunity. The PPI network identified Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interleukin-10 (IL-10), Interleukin-17A (IL-17A), and Interleukin-18 (IL-18) as key proteins. The correlation analysis showed that acetic acid and valerate were closely related to the immune response. In this study, the differences in cecal short-chain fatty acids and the immune barrier between Dahe pigs and Dahe black pigs were compared, which provided a theoretical basis for improving the intestinal immunity of pigs.
Collapse
Affiliation(s)
- Huijin Jia
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yuxiao Xie
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China
| | - Lanlan Yi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wenjie Cheng
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Guangyao Song
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wenzhe Shi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Junhong Zhu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Sumei Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
15
|
Niu X, Chang G, Xu N, Li R, Niu B, Mao R, Wang S, Li G, Jiang J, Wang L. Vitamin A-Integrated Cinnamaldehyde Nanoemulsion: A Nanotherapeutic Approach To Counteract Liver Fibrosis via Gut-Liver Axis Modulation. ACS NANO 2025; 19:10433-10451. [PMID: 40045827 DOI: 10.1021/acsnano.5c00136] [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: 03/19/2025]
Abstract
Liver fibrosis, a complex process resulting from most chronic liver diseases, remains devoid of effective treatments. An increasing body of evidence links liver fibrosis to the "gut-liver axis", with disruptions in the gut microbiota-host balance emerging as a critical contributor to its progression. Cinnamaldehyde (Cin), a natural compound with antioxidant, anti-inflammatory, and anticytotoxic properties, has shown potential in counteracting hepatic stellate cell (HSC) activation. Additionally, Cin has been shown to promote probiotics in the intestine, thereby restoring a healthy microbial community. These characteristics position Cin as a promising candidate for liver fibrosis treatment through modulation of the gut-liver axis. In this study, a Vitamin A (Va)-formulated Cin Nanoemulsion (Va-Cin@NM) was developed to enhance the physicochemical stability of Cin while preserving intestinal homeostasis and facilitating targeted liver deposition. In bile duct ligation (BDL)-induced liver fibrosis in rats, Va-Cin@NM intervention significantly reduced bile duct-like structure proliferation and collagen deposition in the liver. These effects are likely attributed to the restoration of gut microbiota, increased short-chain fatty acid (SCFA) concentrations, and improved intestinal integrity. Moreover, Va-Cin@NM treatment suppressed harmful bacterial populations in the liver, thus mitigating immune injury and inflammatory cell recruitment. Consequently, oxidative stress and HSC activation were attenuated. Overall, Va-Cin@NM demonstrates significant potential as a nanotherapeutic approach for liver fibrosis by modulating the gut-liver axis.
Collapse
Affiliation(s)
- Xia Niu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ge Chang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ning Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Rui Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bingyu Niu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Rui Mao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Guiling Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lulu Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
16
|
Čuljak N, Bendelja K, Leboš Pavunc A, Butorac K, Banić M, Savić Mlakar A, Cvetić Ž, Hrsan J, Novak J, Šušković J, Kos B. In Vitro Analysis of Probiotic Properties Related to the Adaptation of Levilactobacillus brevis to Intestinal Microenvironment and Involvement of S-Layer Proteins. Int J Mol Sci 2025; 26:2425. [PMID: 40141069 PMCID: PMC11942123 DOI: 10.3390/ijms26062425] [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: 02/04/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Although rare, the ability to produce surface S-layer proteins is beneficially associated with particular Lactobacillus strains being investigated as probiotics. Therefore, this work aimed to study specific probiotic functionalities of selected Levilactobacillus brevis strains MB1, MB2, MB13 and MB20, isolated from human milk microbiota, and to assess the contribution of S-proteins. Firstly, Rapid Annotation using Subsystem Technology revealed that cell wall-related genes were abundant in analysed L. brevis genomes. Furthermore, the results demonstrated that S-proteins mediate aggregation capacity and competitive exclusion of selected pathogens by L. brevis strains. The improvement of Caco-2 epithelial monolayer barrier function was demonstrated by the increase in JAM-A and occludin expressions when L. brevis strains or S-proteins were added, with the effect being most pronounced after treatment with MB2 and S-proteins of MB1. L. brevis strains, especially MB20, exerted the potential to adhere to recombinant human ZG16. Strain MB2 and MB20-S-proteins improved the barrier function of HT29 epithelial monolayer, as evidenced by increased ZG16 expression. Analysed L. brevis strains and S-proteins differentially affected the protein expression of IL-1β, IL-6 and IL-8, and IL-10 cytokines. The most prominent effect was observed by S-proteins of MB20, since IL-1β production was decreased while IL-10 production was significantly increased.
Collapse
Affiliation(s)
- Nina Čuljak
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Krešo Bendelja
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Andreja Leboš Pavunc
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Katarina Butorac
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Martina Banić
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Ana Savić Mlakar
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Željko Cvetić
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Jana Hrsan
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Jasna Novak
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Jagoda Šušković
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Blaženka Kos
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| |
Collapse
|
17
|
Zhu S, Yu Q, Xue Y, Li J, Huang Y, Liu W, Wang G, Wang L, Zhai Q, Zhao J, Zhang H, Chen W. Bifidobacterium bifidum CCFM1163 alleviates cathartic colon by activating the BDNF-TrkB-PLC/IP 3 pathway to reconstruct the intestinal nerve and barrier. Food Funct 2025; 16:2057-2072. [PMID: 39963068 DOI: 10.1039/d4fo05835f] [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: 03/04/2025]
Abstract
Introduction: Cathartic colon (CC) is a type of slow-transit constipation caused by a patient's long-term use of irritating laxatives. Probiotics play a crucial role in managing constipation. Objectives: This study aims to identify probiotics that can alleviate CC and explore their specific mechanisms of action. Methods: The CC-model was constructed using senna leaf extract. Bifidobacterium bifidum was applied to the mice for intervention. Relevant marker changes were then examined using ELISA and RT-qPCR. Furthermore, 16S rDNA sequencing was utilized for functional prediction of intestinal microorganisms, while GC-MS analysis was performed to determine the content of short-chain fatty acids (SCFAs) in feces. Results: Senna damages the intestinal nerve and the intestinal barrier while inducing CC. In contrast, Bifidobacterium bifidum CCFM1163 may enhance the brain-derived neurotrophic factor (BDNF) expression in the colon by altering the intestinal microbiota composition (e.g., increasing Lactobacillus and Bacteroides, and decreasing Faecalibaculum) and by elevating SCFA levels (e.g., acetic and isobutyric acid). Subsequently, elevated BDNF expression activates the BDNF-tyrosine kinase receptor B-phospholipase C/inositol trisphosphate (BDNF-TrkB-PLC/IP3) pathway, which upregulates the gene expression of Uchl1, S100β, and Acta2; repairs the enteric nervous system-interstitial cells of Cajal-smooth muscle cells (ENS-ICC-SMC) network; upregulates the gene expression of Ocln and Tjp1; improves intestinal permeability in CC mice; and modulates the immune response by upregulating Tlr4, downregulating Il1b, and upregulating Il10, ultimately alleviating CC. Conclusion: Bifidobacterium bifidum CCFM1163 was identified as a probiotic that can promote BDNF expression in the colon, activate the BDNF-TrkB-PLC/IP3 signaling pathway, and effectively alleviate CC.
Collapse
Affiliation(s)
- Shengnan Zhu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qiangqing Yu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yifan Xue
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jiazhen Li
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yin Huang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenxu Liu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Linlin Wang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
18
|
Jiang J, Xie H, Cao S, Xu X, Zhou J, Liu Q, Ding C, Liu M. Post-stroke depression: exploring gut microbiota-mediated barrier dysfunction through immune regulation. Front Immunol 2025; 16:1547365. [PMID: 40098959 PMCID: PMC11911333 DOI: 10.3389/fimmu.2025.1547365] [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: 12/18/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Post-stroke depression (PSD) is one of the most common and devastating neuropsychiatric complications in stroke patients, affecting more than one-third of survivors of ischemic stroke (IS). Despite its high incidence, PSD is often overlooked or undertreated in clinical practice, and effective preventive measures and therapeutic interventions remain limited. Although the exact mechanisms of PSD are not fully understood, emerging evidence suggests that the gut microbiota plays a key role in regulating gut-brain communication. This has sparked great interest in the relationship between the microbiota-gut-brain axis (MGBA) and PSD, especially in the context of cerebral ischemia. In addition to the gut microbiota, another important factor is the gut barrier, which acts as a frontline sensor distinguishing between beneficial and harmful microbes, regulating inflammatory responses and immunomodulation. Based on this, this paper proposes a new approach, the microbiota-immune-barrier axis, which is not only closely related to the pathophysiology of IS but may also play a critical role in the occurrence and progression of PSD. This review aims to systematically analyze how the gut microbiota affects the integrity and function of the barrier after IS through inflammatory responses and immunomodulation, leading to the production or exacerbation of depressive symptoms in the context of cerebral ischemia. In addition, we will explore existing technologies that can assess the MGBA and potential therapeutic strategies for PSD, with the hope of providing new insights for future research and clinical interventions.
Collapse
Affiliation(s)
- Jia Jiang
- The Second Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Haihua Xie
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Sihui Cao
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Xuan Xu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Jingying Zhou
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Qianyan Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Changsong Ding
- School of Information Science and Engineering, Hunan University of Chinese Medicine, Changsha, China
| | - Mi Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
19
|
Liu Y, Yan D, Chen R, Zhang Y, Wang C, Qian G. Recent insights and advances in gut microbiota's influence on host antiviral immunity. Front Microbiol 2025; 16:1536778. [PMID: 40083779 PMCID: PMC11903723 DOI: 10.3389/fmicb.2025.1536778] [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: 11/29/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
A diverse array of microbial organisms colonizes the human body, collectively known as symbiotic microbial communities. Among the various pathogen infections that hosts encounter, viral infections represent one of the most significant public health challenges worldwide. The gut microbiota is considered an important biological barrier against viral infections and may serve as a promising target for adjuvant antiviral therapy. However, the potential impact of symbiotic microbiota on viral infection remains relatively understudied. In this review, we discuss the specific regulatory mechanisms of gut microbiota in antiviral immunity, highlighting recent advances in how gut microbiota regulate the host immune response, produce immune-related molecules, and enhance the host's defense against viruses. Finally, we also discuss the antiviral potential of oral probiotics.
Collapse
Affiliation(s)
- Ying Liu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Danying Yan
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Ran Chen
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingying Zhang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Chuwen Wang
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Guoqing Qian
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Infectious Diseases, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
20
|
Dong Y, Yang S, Zhang S, Zhao Y, Li X, Han M, Gai Z, Zou K. Modulatory impact of Bifidobacterium longum subsp. longum BL21 on the gut-brain-ovary axis in polycystic ovary syndrome: insights into metabolic regulation, inflammation mitigation, and neuroprotection. mSphere 2025; 10:e0088724. [PMID: 39898662 PMCID: PMC11853005 DOI: 10.1128/msphere.00887-24] [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: 10/22/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
This study evaluates the efficacy of Bifidobacterium longum subsp. longum BL21 in mitigating symptoms of polycystic ovarian syndrome (PCOS) in DHT-induced PCOS model mice. It focuses on BL21's role in modulating metabolic dysregulation, inflammation, and neuroprotection via the gut-brain-ovary axis. Employing an 8-week treatment regimen, this research assessed the effects of BL21 on prenatal androgen-induced PCOS in ICR mice. Evaluations included body weight, glucose tolerance tests, serum analyses of BDNF, inflammatory markers, sex hormone levels, and 16S rRNA gene sequencing for gut microbiota diversity and composition. Twenty-four ICR mice with induced PCOS served as subjects to examine the probiotic's impact. Mice were administered a daily oral dose of 1 × 109 CFU of BL21 continuously for a total of 8 weeks. BL21 significantly enhanced sex hormone levels (P < 0.05), particularly those of follicle-stimulating hormone (FSH) and estradiol (E2), indicating improved ovarian function and offering a novel PCOS treatment approach. The intervention notably curbed weight gain and improved glucose tolerance in PCOS mice (P < 0.05). BL21 reduced inflammatory markers such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and lipopolysaccharides (LPS), while increasing IL-10, BDNF, FSH, and E2 levels (P < 0.05 for all). It also enriched gut microbiota diversity, enhancing populations of Bifidobacterium and Lactobacillus. Correlation analyses underscored the positive shifts in microbiota linked to beneficial hormonal and inflammatory profiles. BL21 shows promise in alleviating PCOS symptoms through metabolic regulation, inflammation reduction, and neuroprotection, validating its potential in integrated therapeutic strategies.IMPORTANCEPolycystic ovarian syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, characterized by metabolic irregularities, hormonal imbalances, and chronic inflammation. Existing treatments are often inadequate, addressing symptoms without targeting the underlying etiological factors. The investigation of Bifidobacterium longum subsp. longum BL21 as a probiotic intervention offers a novel approach by potentially regulating the gut-brain-ovary axis. This could lead to innovative therapeutic strategies that not only manage but also potentially reverse the multifaceted symptoms of PCOS, enhancing quality of life and reproductive health.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Shengnan Yang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Shu Zhang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Yuan Zhao
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xinlan Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Mei Han
- Department of Food Science, Shanghai Business School, Shanghai, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
21
|
Zhang X, Cui Y, Zhang Z, Huang X, Zhang X, Hu X, Li T, Li S. Effects of hawthorn pectin and its oligomers on gut microbiota and metabolites in high-fat diet mice. Food Funct 2025; 16:1205-1217. [PMID: 39807952 DOI: 10.1039/d4fo04686b] [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: 01/16/2025]
Abstract
Pectin is an acidic heteropolysaccharide with natural, green, and inexpensive characteristics. Compared to polysaccharides, oligosaccharides are more easily utilized by the body, and the physiological function of hawthorn pectin oligosaccharides (POS) may vary depending on their degree of polymerization (DP). Therefore, we mainly studied the effects of hawthorn pectin (HP) and POS with different DP on gut microbiota disorders induced by high-fat diet (HFD). HP and POS both improved weight gain, dyslipidemia, and glucose homeostasis caused by HFD, and increased serum GLP-1 levels. Meanwhile, the increased expression of Gcg and Pcsk1 genes in the ileum of the treatment group further confirmed this result. In addition, HP and POS reduced certain opportunistic pathogens, while restoring the richness and diversity of the gut microbiota. Meanwhile, HP and POS can improve intestinal barrier dysfunction by increasing the claudin-1, occludin, ZO-1, and MUC2 genes. Furthermore, fecal metabolomics suggests that POS may enhance linoleic acid synthesis and improve lipid metabolism by upregulating 9,10-DHOME ((12Z)-9,10-dihydroxyoctadec-12-enoic acid), while HP cannot. Overall, the research results indicate that both HP and POS can improve the weight phenotype changes, gut microbiota disruption, and metabolites changes caused by HFD. Particularly, POS has a better effect than HP, and there are differences in the improvement effect of POS with different DP, among which POS with DP 5 has the most significant improvement effect. This discovery enhances a deeper comprehension of the biological activity of different POS, providing an important basis for further optimizing the application of POS as a functional food.
Collapse
Affiliation(s)
- Xiushan Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Yanmin Cui
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Zuoyi Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Xin Huang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Xiaowei Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Xiaopei Hu
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Tuoping Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| | - Suhong Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China.
| |
Collapse
|
22
|
Yang F, Li X, Sun J, Pang X, Sun Q, Lu Y. Regulatory mechanisms of the probiotic-targeted gut-liver axis for the alleviation of alcohol-related liver disease: a review. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 39905925 DOI: 10.1080/10408398.2025.2455954] [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: 02/06/2025]
Abstract
Alcohol abuse-triggered alcohol-related liver disease (ALD) has become as a global public health concern that substantially affects the well-being and clinical status of patients. Although modern medicine provides various treatments for ALD, their effectiveness is limited and can lead to adverse side effects. Probiotics have been employed to prevent, alleviate, and even treat ALD, with promising results. However, few comprehensive reviews are available on how they mitigate ALD by targeting the gut-liver axis. This review systematically clarifies the specific mediators of the gut-liver axis in healthy states. It also describes the alterations observed in ALD. Furthermore, this review thoroughly summarizes the underlying mechanisms through which probiotics act on the gut-liver axis to relieve ALD. It also discusses the current status and challenges faced in clinical research applications. Finally, we discuss the challenges and future prospects of using probiotics to treat ALD. This review improves our understanding of ALD and supports the development and application of probiotics that target the gut-liver axis for therapeutic use.
Collapse
Affiliation(s)
- Feiyu Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
23
|
Chen A, Gong Y, Wu S, Du Y, Liu Z, Jiang Y, Li J, Miao YB. Navigating a challenging path: precision disease treatment with tailored oral nano-armor-probiotics. J Nanobiotechnology 2025; 23:72. [PMID: 39893419 PMCID: PMC11786591 DOI: 10.1186/s12951-025-03141-3] [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: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Oral probiotics have significant potential for preventing and treating many diseases. Yet, their efficacy is often hindered by challenges related to survival and colonization within the gastrointestinal tract. Nanoparticles emerge as a transformative solution, offering robust protection and enhancing the stability and bioavailability of these probiotics. This review explores the innovative application of nanoparticle-armored engineered probiotics for precise disease treatment, specifically addressing the physiological barriers associated with oral administration. A comprehensive evaluation of various nano-armor probiotics and encapsulation methods is provided, carefully analyzing their respective merits and limitations, alongside strategies to enhance probiotic survival and achieve targeted delivery and colonization within the gastrointestinal tract. Furthermore, the review explores the potential clinical applications of nano-armored probiotics in precision therapeutics, critically addressing safety and regulatory considerations, and proposing the innovative concept of 'probiotic intestinal colonization with nano armor' for brain-targeted therapies. Ultimately, this review aspires to guide the advancement of nano-armored probiotic therapies, driving progress in precision medicine and paving the way for groundbreaking treatment modalities.
Collapse
Affiliation(s)
- Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Shaoquan Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ye Du
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Jiahong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| |
Collapse
|
24
|
Li J, Luo T, Wang D, Zhao Y, Jin Y, Yang G, Zhang X. Therapeutic application and potential mechanism of plant-derived extracellular vesicles in inflammatory bowel disease. J Adv Res 2025; 68:63-74. [PMID: 38341033 PMCID: PMC11785581 DOI: 10.1016/j.jare.2024.01.035] [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: 10/18/2023] [Revised: 01/09/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Plant-derived extracellular vesicles (PDEVs) are membrane vesicles characterized by a phospholipid bilayer as the basic skeleton that is wrapped by various functional components of proteins and nucleic acids. An increasing number of studies have confirmed that PDEVs can be a potential treatment of inflammatory bowel disease (IBD) and can, to some extent, compensate for the limitations of existing therapies. AIM OF REVIEW This review summarizes the recent advances and potential mechanisms underlying PDEVs obtained from different sources to alleviate IBD. In addition, the review discusses the possible applications and challenges of PDEVs, providing a theoretical basis for exploring novel and practical therapeutic strategies for IBD. KEY SCIENTIFIC CONCEPTS OF REVIEW In IBD, the crosstalk mechanism of PDEVs may regulate the intestinal microenvironment homeostasis, especially immune responses, the intestinal barrier, and the gut microbiota. In addition, drug loading enhances the therapeutic potential of PDEVs, particularly regarding improved tissue targeting and stability. In the future, not only immunotherapy based on PDEVs may be an effective treatment for IBD, but also the intestinal barrier and intestinal microbiota will be a new direction for the treatment of IBD.
Collapse
Affiliation(s)
- Jinling Li
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, Zhejiang Province, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Ting Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Yao Zhao
- Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China
| | - Yuanxiang Jin
- Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang Province, China
| | - Guiling Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China; Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China.
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, Zhejiang Province, China.
| |
Collapse
|
25
|
Habibi A, Letafatkar N, Sattari N, Nobakht S, Rafat Z, Soltani Moghadam S, Mirdamadi A, Javid M, Jamilian P, Hassanipour S, Keivanlou MH, Amini-Salehi E. Modulation of inflammatory markers in type 2 diabetes mellitus through gut microbiome-targeted interventions: An umbrella review on meta-analyses. Clin Nutr ESPEN 2025; 65:93-104. [PMID: 39551350 DOI: 10.1016/j.clnesp.2024.11.011] [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: 05/07/2024] [Revised: 09/23/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND & AIMS Type 2 diabetes mellitus (T2DM) poses a significant global health challenge due to various lifestyle factors contributing to its prevalence and associated complications. Chronic low-grade inflammation, characterized by elevated levels of inflammatory markers such as C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α), plays a pivotal role in the pathogenesis of T2DM. Modulation of the gut microbiota through microbiome-targeted therapy (MTT), including probiotics, prebiotics, and synbiotics, has emerged as a potential strategy to mitigate inflammation and improve metabolic outcomes in T2DM. METHODS A systematic review and meta-analysis were conducted following PRISMA guidelines to evaluate the impact of MTT on inflammatory markers in patients with T2DM. Searches were performed in PubMed, Scopus, and Web of Science databases up to June 2024, with inclusion criteria limited to English-language meta-analyses of randomized controlled trials (RCTs) assessing the effects of probiotics, prebiotics, or synbiotics on inflammatory markers in T2DM patients. RESULTS Ten meta-analyses met the inclusion criteria, comprising studies investigating the effects of various MTT interventions on CRP, IL-6, and TNF-α levels in T2DM patients. Meta-analysis results indicated significant reductions in CRP (SMD: -0.070; 95 % CI: -0.119 to -0.020) and TNF-α (SMD: -0.370; 95 % CI: -0.554 to -0.186) levels following MTT, while IL-6 reductions (SMD: -0.070; 95 % CI: -0.269 to 0.129) did not reach statistical significance. However, heterogeneity in study quality, intervention protocols, and participant demographics posed challenges in interpretation. CONCLUSIONS While improvements in inflammatory markers with MTT have been observed, significant limitations-such as heterogeneity in study quality and variation in intervention protocols-highlight the need for further research to confirm its efficacy and clarify underlying mechanisms. Future studies should aim to address these limitations by exploring variations in dosage, supplement formulations, and bacterial strains, which are crucial for improving the reliability and broader applicability of MTT in the management of T2DM.
Collapse
Affiliation(s)
- Arman Habibi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Negin Letafatkar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sara Nobakht
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Rafat
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Arian Mirdamadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mona Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mohammad-Hossein Keivanlou
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
26
|
Mafe AN, Iruoghene Edo G, Akpoghelie PO, Gaaz TS, Yousif E, Zainulabdeen K, Isoje EF, Igbuku UA, Opiti RA, Garba Y, Essaghah AEA, Ahmed DS, Umar H. Probiotics and Food Bioactives: Unraveling Their Impact on Gut Microbiome, Inflammation, and Metabolic Health. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10452-2. [PMID: 39808399 DOI: 10.1007/s12602-025-10452-2] [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: 01/03/2025] [Indexed: 01/16/2025]
Abstract
This review paper delves into the role of probiotics and food bioactives in influencing gut health and overall well-being, within the context of probiotics and food bioactives, emphasizing their roles in modulating inflammation, gut microbiota, and metabolic health. Probiotics are defined as live microorganisms that confer health benefits to the host, primarily through their impact on the gut microbiome; a complex community of microorganisms crucial for maintaining health. The review aims to elucidate how probiotics, incorporated into both traditional and modern food systems, can enhance gut health and address metabolic disorders. It examines the types of probiotics present in various foods and their mechanisms of action, including their effects on immune function and metabolic health. By exploring the links between probiotics and health outcomes such as digestive health, immune support, and mental health, the review identifies specific conditions where probiotics show significant promise. Hurldes such as inconsistencies in research findings, variability in probiotic strains, and dosages are addressed. The paper also suggests future research directions, including the potential for personalized probiotic interventions. The review concludes by summarizing key findings and emphasizing the critical role of probiotics in food systems for promoting overall health and mitigating metabolic diseases.
Collapse
Affiliation(s)
- Alice Njolke Mafe
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Jalingo, Taraba State, Nigeria
| | - Great Iruoghene Edo
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria.
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq.
| | - Patrick Othuke Akpoghelie
- Department of Food Science and Technology, Faculty of Science, Delta State University of Science and Technology, Ozoro, Delta State, Nigeria
| | - Tayser Sumer Gaaz
- Department of Prosthetics and Orthotics Engineering, College of Engineering and Technologies, Al-Mustaqbal University, Babylon, Iraq
| | - Emad Yousif
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Khalid Zainulabdeen
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Endurance Fegor Isoje
- Department of Science Laboratory Technology (Biochemistry Option), Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Ufuoma Augustina Igbuku
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Rapheal Ajiri Opiti
- Department of Petroleum Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Yasal Garba
- Department of Information Engineering, College of Information Engineering, Al-Nahrain University, Baghdad, Iraq
| | - Arthur Efeoghene Athan Essaghah
- Department of Urban and Regional Planning, Faculty of Environmental Sciences, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Dina S Ahmed
- Department of Chemical Industries, Institute of Technology-Baghdad, Middle Technical University, Baghdad, Iraq
| | - Huzaifa Umar
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus
| |
Collapse
|
27
|
Chowdhury MR, Islam A, Yurina V, Shimosato T. Water pollution, cholera, and the role of probiotics: a comprehensive review in relation to public health in Bangladesh. Front Microbiol 2025; 15:1523397. [PMID: 39877756 PMCID: PMC11772269 DOI: 10.3389/fmicb.2024.1523397] [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: 11/06/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Cholera, a disease caused by Vibrio cholerae, remains a pervasive public health threat, particularly in regions with inadequate water sanitation and hygiene infrastructure, such as Bangladesh. This review explores the complex interplay between water pollution and cholera transmission in Bangladesh, highlighting how contaminated water bodies serve as reservoirs for V. cholerae. A key focus is the potential role of probiotics as a novel intervention approach for cholera prevention and management. Probiotics are promising as an adjunctive approach to existing therapies as they can enhance gut barrier function, induce competitive exclusion of pathogens, and modulate host immune responses. Recent probiotic advancements include engineering strains that disrupt V. cholerae biofilms and inhibit their virulence. Integrating probiotics with traditional cholera control measures could significantly enhance their effectiveness and provide a multifaceted approach to combating this persistent disease. This review aims to shed light on the potential of probiotics in revolutionizing cholera management and to offer insights into their application as both preventive and therapeutic tools in the fight against this enduring public health challenge.
Collapse
Affiliation(s)
- Md. Rayhan Chowdhury
- Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
| | - Ariful Islam
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Valentina Yurina
- Department of Pharmacy, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Takeshi Shimosato
- Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
- Department of Pharmacy, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Institute for Aqua Regeneration, Shinshu University, Nagano, Japan
| |
Collapse
|
28
|
Gómez-Velázquez HDJ, González-Dávalos L, de los Ríos EA, Figueroa-Cárdenas JDD, Vázquez-Durán A, Méndez-Albores A, Shimada A, Mora O. Physicochemical characterization and 16S rRNA analysis of a direct-fed microbial from calf ruminal fluid and its protective effect on Sprague-Dawley rat gut barrier function. Transl Anim Sci 2025; 9:txaf003. [PMID: 40083360 PMCID: PMC11905223 DOI: 10.1093/tas/txaf003] [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: 11/05/2024] [Accepted: 01/08/2025] [Indexed: 03/16/2025] Open
Abstract
This study aimed to characterize the physicochemical properties and microbiota composition of a direct-fed microbial (DFM) and evaluate its protective effect on intestinal permeability in Sprague-Dawley rats using fluorescein isothiocyanate dextran (FITC-d) as a biomarker. The DFM was further characterized using Fourier-transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), environmental scanning electron microscopy (ESEM), energy-dispersive X-ray spectroscopy (EDS), and cell surface hydrophobicity (microbial adhesion to hexadecane [MATH] assay). The 16S rRNA gene was sequenced using short-read sequencing. In general, the DFM exhibited the characteristic FTIR bands associated with probiotic cells with a protein/carbohydrate ratio of 1.3:1. It was also found from the DLS analysis that the average particle size and surface electrical potential of the probiotic cells were 1,062 ± 77 nm and -32.6 ± 3.7 mV, respectively. ESEM studies confirmed the size of the cells (1,010 to 1,060 nm), showing a quasi-spherical cocci-type morphology, whereas EDS spectroscopy revealed a higher Nitrogen/Carbone ratio on the cell surface. Moreover, the MATH assay showed the hydrophobic character of the DFM (92% adhesion). Furthermore, based on the 16S rRNA gene analysis, the predominant genus in the DFM was Streptococcus (99%). Regarding the protective effect on the gut barrier, animals supplemented with 1011 CFU/mL exhibited a significantly reduced intestinal permeability compared with the control group. DFM supplementation also increased villi and crypt dimensions and Goblet cells (P < 0.05) in the ileum and cecum. These results demonstrate that the DFM presented adequate surface and colloidal properties that help maintain the functionality of the gut barrier.
Collapse
Affiliation(s)
- Haiku D J Gómez-Velázquez
- Laboratorio de Rumiología y Metabolismo Nutricional (RuMeN), Facultad de Estudios Superiores-Cuautitlán (FESC), Universidad Nacional Autónoma de México (UNAM), Querétaro, Querétaro, Mexico
| | - Laura González-Dávalos
- Laboratorio de Rumiología y Metabolismo Nutricional (RuMeN), Facultad de Estudios Superiores-Cuautitlán (FESC), Universidad Nacional Autónoma de México (UNAM), Querétaro, Querétaro, Mexico
| | - Erika A de los Ríos
- Unidad de Microscopía, Instituto de Neurobiología, UNAM, Querétaro, Querétaro, México
| | - Juan de Dios Figueroa-Cárdenas
- Materiales Bio-orgánicos, CINVESTAV-Unidad Querétaro, Libramiento Norponiente No. 2000, Fraccionamiento Real de Juriquilla, Querétaro, Querétaro, México
| | - Alma Vázquez-Durán
- Ciencia y Tecnología de Materiales, Unidad de Investigación Multidisciplinaria L14-A1 (Ciencia y Tecnología de Materiales), FESC, UNAM, Cuautitlán Izcalli, Estado de México, México
| | - Abraham Méndez-Albores
- Ciencia y Tecnología de Materiales, Unidad de Investigación Multidisciplinaria L14-A1 (Ciencia y Tecnología de Materiales), FESC, UNAM, Cuautitlán Izcalli, Estado de México, México
| | - Armando Shimada
- Laboratorio de Rumiología y Metabolismo Nutricional (RuMeN), Facultad de Estudios Superiores-Cuautitlán (FESC), Universidad Nacional Autónoma de México (UNAM), Querétaro, Querétaro, Mexico
| | - Ofelia Mora
- Laboratorio de Rumiología y Metabolismo Nutricional (RuMeN), Facultad de Estudios Superiores-Cuautitlán (FESC), Universidad Nacional Autónoma de México (UNAM), Querétaro, Querétaro, Mexico
| |
Collapse
|
29
|
Huang H, Zhao L, Kong X, Zhu J, Lu J. Vinegar powder exerts immunomodulatory effects through alleviating immune system damage and protecting intestinal integrity and microbiota homeostasis. FOOD BIOSCI 2025; 63:105687. [DOI: 10.1016/j.fbio.2024.105687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Montanari M, Mercuri NB, Martella G. Exceeding the Limits with Nutraceuticals: Looking Towards Parkinson's Disease and Frailty. Int J Mol Sci 2024; 26:122. [PMID: 39795979 PMCID: PMC11719863 DOI: 10.3390/ijms26010122] [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: 11/16/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
One of the most pressing challenges facing society today is the rising prevalence of physical and cognitive frailty. This geriatric condition makes older adults more vulnerable to disability, illness, and a heightened risk of mortality. In this scenario, Parkinson's disease (PD) and geriatric frailty, which share several common characteristics, are becoming increasingly prevalent worldwide, underscoring the urgent need for innovative strategies. Nutraceuticals are naturally occurring bioactive compounds contained in foods, offering health benefits over and above essential nutrition. By examining the literature from the past decade, this review highlights how nutraceuticals can act as complementary therapies, addressing key processes, such as oxidative stress, inflammation, and neuroprotection. Notably, the antioxidant action of nutraceuticals appears particularly beneficial in regard to PD and geriatric frailty. For instance, antioxidant-rich nutraceuticals may mitigate the oxidative damage linked to levodopa therapy in PD, potentially reducing the side effects and enhancing treatment sustainability. Similarly, the antioxidant effects of nutraceuticals may amplify the benefits of physical activity, enhancing muscle function, cognitive health, and resilience, thereby reducing the risk of frailty. This review proposes a holistic approach integrating nutraceuticals with exercise, pharmacotherapy, and lifestyle adjustments. It promises to transform the management of ARD, prolong life, and improve the quality of life and well-being of older people.
Collapse
Affiliation(s)
- Martina Montanari
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy;
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
- Department of Wellbeing, Nutrition and Sport, Faculty of Humanities Educations and Sports, Pegaso Telematics University, 80145 Naples, Italy
| |
Collapse
|
31
|
Tsuji K, Uchida N, Nakanoh H, Fukushima K, Haraguchi S, Kitamura S, Wada J. The Gut-Kidney Axis in Chronic Kidney Diseases. Diagnostics (Basel) 2024; 15:21. [PMID: 39795549 PMCID: PMC11719742 DOI: 10.3390/diagnostics15010021] [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: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
The gut-kidney axis represents the complex interactions between the gut microbiota and kidney, which significantly impact the progression of chronic kidney disease (CKD) and overall patient health. In CKD patients, imbalances in the gut microbiota promote the production of uremic toxins, such as indoxyl sulfate and p-cresyl sulfate, which impair renal function and contribute to systemic inflammation. Mechanisms like endotoxemia, immune activation and oxidative stress worsen renal damage by activating pro-inflammatory and oxidative pathways. Insights into these mechanisms highlight the impact of gut-derived metabolites, bacterial translocation, and immune response changes on kidney health, suggesting new potential approaches for CKD treatment. Clinical applications, such as dietary interventions, prebiotics, probiotics and fecal microbiota transplantation, are promising in adjusting the gut microbiota to alleviate CKD symptoms and slow disease progression. Current research highlights the clinical relevance of the gut-kidney axis, but further study is essential to clarify these mechanisms' diagnostic biomarkers and optimize therapeutic interventions. This review emphasizes the importance of an integrated approach to CKD management, focusing on the gut microbiota as a therapeutic target to limit kidney injury.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Naruhiko Uchida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Soichiro Haraguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nephrology, Aoe Clinic, Okayama 700-8607, Japan
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| |
Collapse
|
32
|
Barbosa AMS, Carvalho MPS, Naves LDP, da Motta SAB, Chaves RF, Resende M, Lima DD, Hansen LHB, Cantarelli VDS. Performance and Health Parameters of Sows and Their Litters Using a Probiotic Supplement Composed of Bacillus subtilis 541 and Bacillus amyloliquefaciens 516. Animals (Basel) 2024; 14:3511. [PMID: 39682476 DOI: 10.3390/ani14233511] [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: 09/25/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
This study investigated the efficacy of using probiotics on the performance and health parameters of sows and their litters. A randomized block design was used with 584 sows and 292 replications, with two dietary treatments: the control group (basal diet without probiotics) and the probiotic group (basal diet supplemented with 400 g/ton of a probiotic composed of Bacillus subtilis (B. subtilis) 541 and Bacillus amyloliquefaciens (B. amyloliquefaciens) 516). Feed intake was evaluated throughout the experimental period. Bodyweight and backfat thickness of the sows were measured at the beginning and end of each phase. Piglets were weighed individually at birth and at weaning. Performance variables and physiological parameters were analyzed. Sows that received the probiotic supplement exhibited increased milk production (p = 0.05) and bodyweight loss, along with reduced postpartum cortisol levels (p < 0.05). The piglets from the probiotic treatment group had higher (p < 0.001) weaning weight and fewer (p < 0.05) crushing deaths, received fewer (p < 0.001) medications, and had lower (p < 0.05) excretion of pathogenic bacteria and lower (p < 0.05) excretion of fecal Lactobacillus sp. They also had higher (p < 0.05) concentration of fecal myeloperoxidase (MPO) close to weaning and improved ileal histomorphometric measures. In conclusion, supplementation with the probiotic product improves performance and promotes health parameters of the sows their litters.
Collapse
Affiliation(s)
- Aline Maria Silva Barbosa
- Faculty of Medicine Veterinary and Animal Science, Federal University of Lavras, Lavras 37200-000, MG, Brazil
| | - Maria Paula Souza Carvalho
- Faculty of Medicine Veterinary and Animal Science, Federal University of Lavras, Lavras 37200-000, MG, Brazil
| | - Luciana de Paula Naves
- Faculty of Medicine Veterinary and Animal Science, Federal University of Lavras, Lavras 37200-000, MG, Brazil
| | | | - Rhuan Filipe Chaves
- Animalnutri Ciência e Tecnologia, José de Santana, Centro, 520, Patos de Minas 38700-052, MG, Brazil
| | - Maíra Resende
- Animalnutri Ciência e Tecnologia, José de Santana, Centro, 520, Patos de Minas 38700-052, MG, Brazil
| | | | | | | |
Collapse
|
33
|
Seo H, Kim S, Beck S, Song HY. Perspectives on Microbiome Therapeutics in Infectious Diseases: A Comprehensive Approach Beyond Immunology and Microbiology. Cells 2024; 13:2003. [PMID: 39682751 PMCID: PMC11640688 DOI: 10.3390/cells13232003] [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: 11/02/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Although global life expectancy has increased over the past 20 years due to advancements in managing infectious diseases, one-fifth of people still die from infections. In response to this ongoing threat, significant efforts are underway to develop vaccines and antimicrobial agents. However, pathogens evolve resistance mechanisms, complicating their control. The COVID-19 pandemic has underscored the limitations of focusing solely on the pathogen-killing strategies of immunology and microbiology to address complex, multisystemic infectious diseases. This highlights the urgent need for practical advancements, such as microbiome therapeutics, that address these limitations while complementing traditional approaches. Our review emphasizes key outcomes in the field, including evidence of probiotics reducing disease severity and insights into host-microbiome crosstalk that have informed novel therapeutic strategies. These findings underscore the potential of microbiome-based interventions to promote physiological function alongside existing strategies aimed at enhancing host immune responses and pathogen destruction. This narrative review explores microbiome therapeutics as next-generation treatments for infectious diseases, focusing on the application of probiotics and their role in host-microbiome interactions. While offering a novel perspective grounded in a cooperative defense system, this review also addresses the practical challenges and limitations in translating these advancements into clinical settings.
Collapse
Affiliation(s)
- Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Sukyung Kim
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Samuel Beck
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, J-607, 609 Albany, Boston, MA 02118, USA
| | - Ho-Yeon Song
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Cheonan-si 31151, Chungnam-do, Republic of Korea
| |
Collapse
|
34
|
Sivri D, Şeref B, Şare Bulut M, Gezmen Karadağ M. Evaluation of the Effect of Probiotic Supplementation on Intestinal Barrier Integrity and Epithelial Damage in Colitis Disease: A Systematic Review. Nutr Rev 2024:nuae180. [PMID: 39602817 DOI: 10.1093/nutrit/nuae180] [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: 11/29/2024] Open
Abstract
CONTEXT Previous reviews have focused on the effects of probiotics on colitis, but there is a need to understand their impact on barrier integrity and tight junction protein improvement in colitis. OBJECTIVE This study aimed to systematically examine the effects of probiotic use on barrier integrity in colitis disease. This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. DATA SOURCES A systematic search in PubMed, Web of Science, Scopus, and Cochrane databases identified 2537 articles. DATA EXTRACTION As a result of the search, 2537 articles were accessed. Study results were summarized descriptively through discussions by intervention conditions, study population, measurement methods, and key findings. The included studies were independently reviewed and all authors reached consensus on the quality and major findings from the included articles. Forty-six studies that met the inclusion criteria were analyzed within the scope of the systematic review. RESULTS Although the study primarily utilized probiotics from the Lactobacillaceae family (notably, L casei, L reuteri, L rhamnosus, L plantarum, and L pentosus) and the Bifidobacteriaceae family (notably, B breve, B animalis, and B dentium), other probiotics also demonstrated positive effects on tight junction proteins. These effects are attributed to the production of bioactive and metabolic compounds, as well as short-chain fatty acids, which combat pathogens and reduce anti-inflammatory agents. However, it was observed that the effects of these probiotics on tight junction proteins varied depending on the strain and dose. CONCLUSION The beneficial effects of probiotics on remission in inflammatory bowel disease are well documented. Studies show that probiotics generally improve intestinal barrier function, but factors such as dose, duration, and bacterial species combinations need further clarification. Additionally, comprehensive studies are needed to understand how improved barrier function affects absorption in individuals. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023452774.
Collapse
Affiliation(s)
- Dilek Sivri
- Department of Nutrition and Dietetics, Anadolu University, Eskişehir, Türkiye
| | - Betül Şeref
- Department of Nutrition and Dietetics, Karamanoğlu Mehmetbey University, Karaman, Türkiye
| | - Melike Şare Bulut
- Department of Nutrition and Dietetics, Biruni University, Istanbul, Türkiye
| | | |
Collapse
|
35
|
Barache N, Belguesmia Y, Zeghbib W, Ladjouzi R, Ouarabi L, Boudjouan F, Zidi G, Bendali F, Drider D. Characterization and Biological In Vitro Screening of Probiotic Yeast Strains Isolated from Algerian Fruits. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10389-y. [PMID: 39531150 DOI: 10.1007/s12602-024-10389-y] [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: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Interest in Saccharomyces and non-Saccharomyces yeasts as biotechnological agents is growing worldwide. Here, Kluyveromyces marxianus GBC2 and two Saccharomyces cerevisiae strains FBZ4 and FBK9 were isolated from pomegranate (Punica granatum) and fig (Ficus carica), respectively, and extensively characterized for their probiotic attributes and health benefits. Overall, these strains were found to be γ-hemolytic, non-cytotoxic against Caco-2 cells, and sensitive to therapeutic antifungals. In terms of probiotic characterization, the strains were able to survive at pH 2 and in 1% bile and had high hydrophobicity and self-aggregation properties, which could explain their ability to form biofilm on a polystyrene and adhere to Caco-2 cells. Adhesion rates of 23.52%, 14.05%, and 9.44% were recorded at 37 °C for K. marxianus GBC2, S. cerevisiae FBK9, and S. cerevisiae FBZ4, respectively. Furthermore, biological screening showed a cholesterol assimilation of 54.32% for K. marxianus GBC2 and almost 33% for both Saccharomyces, more than 73% α-amylase inhibition, and good antioxidant potential for all strains; however, only K. marxianus GBC2 showed antibacterial activity against Staphylococcus aureus ATCC 25923. In light of these findings, the strains could be potential candidates for the development of novel functional foods and for probiotic applications.
Collapse
Affiliation(s)
- Nacim Barache
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Laboratoire de Microbiologie Appliquée, 06000, Bejaia, Algeria.
| | - Yanath Belguesmia
- UMR Transfrontalière BioEcoAgro INRAe 1158, Université de Lille, 59000, Lille, France
| | - Walid Zeghbib
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Laboratoire de Biochimie Appliquée, 06000, Bejaia, Algeria
| | - Rabia Ladjouzi
- UR DYNAMYC 7380, Faculté de Santé, Université Paris-Est Créteil, Ecole Nationale Vétérinaire d'Alfort, USC Anses, 94010, Créteil, France
| | - Liza Ouarabi
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Laboratoire de Microbiologie Appliquée, 06000, Bejaia, Algeria
| | - Farés Boudjouan
- Université de Bejaia, Faculté de Technologie, Laboratoire de Génie de L'Environnement, 06000, Bejaia, Algeria
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Département de Biotechnologie, 06000, Bejaia, Algeria
| | - Ghania Zidi
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Laboratoire de Microbiologie Appliquée, 06000, Bejaia, Algeria
| | - Farida Bendali
- Université de Bejaia, Faculté Des Sciences de La Nature Et de La Vie, Laboratoire de Microbiologie Appliquée, 06000, Bejaia, Algeria
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro INRAe 1158, Université de Lille, 59000, Lille, France.
| |
Collapse
|
36
|
Yu X, Xiong T, Yu L, Liu G, Yang F, Li X, Wei Y, Wang X, Wei S, Jiang Y, Kong X, Ren S, Shi Y. Gut microbiome and metabolome profiling in coal workers' pneumoconiosis: potential links to pulmonary function. Microbiol Spectr 2024; 12:e0004924. [PMID: 39283109 PMCID: PMC11537036 DOI: 10.1128/spectrum.00049-24] [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: 02/05/2024] [Accepted: 07/02/2024] [Indexed: 11/07/2024] Open
Abstract
Coal workers' pneumoconiosis (CWP) is a severe occupational disease resulting from prolonged exposure to coal dust. However, its pathogenesis remains elusive, compounded by a lack of early detection markers and effective treatments. Although the impact of gut microbiota on lung diseases is acknowledged, its specific role in CWP is unclear. This study aims to explore changes in the gut microbiome and metabolome in CWP, while also assessing the correlation between gut microbes and alterations in lung function. Fecal specimens from 43 CWP patients and 48 dust-exposed workers (DEW) were examined using 16S rRNA gene sequencing for microbiota and liquid chromatography-mass spectrometry for metabolite profiling. We observed similar gut microbial α-diversity but significant differences in flora composition (β-diversity) between patients with CWP and the DEW group. After adjusting for age using multifactorial linear regression analysis (MaAsLin2), the distinct gut microbiome profile in CWP patients revealed an increased presence of pro-inflammatory microorganisms such as Klebsiella and Haemophilus. Furthermore, in CWP patients, alterations in gut microbiota-particularly reduced α-diversity and changes in microbial composition-were significantly correlated with impaired pulmonary function, a relationship not observed in DEW. This underscores the specific impact of gut microbiota on pulmonary health in individuals with CWP. Metabolomic analysis of fecal samples from CWP patients and DEW identified 218 differential metabolites between the two groups, with a predominant increase in metabolites in CWP patients, suggesting enhanced metabolic activity in CWP. Key altered metabolites included various lipids, amino acids, and organic compounds, with silibinin emerging as a potential biomarker. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis linked these metabolites to pathways relevant to the development of pulmonary fibrosis. Additionally, studies on the interaction between microbiota and metabolites showed positive correlations between certain bacteria and increased metabolites in CWP, further elucidating the complex interplay in this disease state. Our findings suggest a potential contributory role of gut microbiota in CWP pathogenesis through metabolic regulation, with implications for diagnostic biomarkers and understanding disease mechanisms, warranting further molecular investigation. IMPORTANCE The findings have significant implications for the early diagnosis and treatment of coal workers' pneumoconiosis, highlighting the potential of gut microbiota as diagnostic biomarkers. They pave the way for new research into gut microbiota-based therapeutic strategies, potentially focusing on modifying gut microbiota to mitigate disease progression.
Collapse
Affiliation(s)
- Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Tao Xiong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Lu Yu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaisheng Liu
- Quality Control Office, Xishan Occupational Disease Prevention and Control Institute, Taiyuan, China
| | - Fan Yang
- Quality Control Office, Xishan Occupational Disease Prevention and Control Institute, Taiyuan, China
| | - Xueqin Li
- Department of Respiratory Medicine, Jincheng General Hospital, Shanxi, China
| | - Yangyang Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojing Wang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuting Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yi Jiang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaomei Kong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shouan Ren
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yiwei Shi
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
37
|
More K, Hanumantharaju A, Amrit A, Nimbalkar SM, Patole S. Use of Probiotics for Preventing Necrotizing Enterocolitis in Preterm Infants: A Survey of Current Practices Among Indian Neonatologists. Cureus 2024; 16:e73923. [PMID: 39697933 PMCID: PMC11654888 DOI: 10.7759/cureus.73923] [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: 11/17/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE Probiotics are known to reduce the risk of necrotizing enterocolitis (NEC≥ Stage II) significantly, as well as all-cause mortality, late-onset sepsis (LOS), and feeding intolerance in preterm infants. Probiotics have been reported to have comparable benefits in high- and low-middle-income countries (LMICs). We aimed to assess the current practices of neonatologists in India for using probiotics in preterm infants. MATERIAL AND METHODS A questionnaire created using Survey Monkey's web-based tool was sent to neonatologists in India. Survey forms automatically converted responses into Excel files (Microsoft® Corp., Redmond, WA). Data were analyzed using SPSS (IBM Corp., Armonk, NY). RESULTS A total of 615 responses were received from various neonatal intensive care units (NICUs) in India (Level I: 43 (7%), II: 124 (20.8%), III: 448 (72.8%)). Around 431 (70%) of the units had either National Neonatology Forum (NNF) accreditation or IAP fellowships or were affiliated with private or government medical colleges. The remaining 184 (30%) were in private setups. Routine probiotic supplementation (RPS) was provided in 241 (39.1%) of the responding units; 179 (48%) quoted inadequate evidence as the reason for not providing RPS, 125 (33.43%) quoted difficulty in sourcing safe and effective products, whereas others were concerned about adverse effects. Most centers provided RPS for preterm infants <32 weeks and 1500 g at birth. The clinical practice was influenced by the judgment of the attending clinician. Significant variation was noticed in the protocol for RPS. CONCLUSION Findings of the survey suggest that approximately 39% of the participating neonatologists in India currently offer RPS for preterm infants. A significant variation exists in the selection of probiotic strains, products, dose, and duration of supplementation. Despite limitations, our findings are useful in guiding clinical practice and further research to optimize the safety and efficacy of RPS for preterm infants.
Collapse
Affiliation(s)
- Kiran More
- Neonatology, MRR Children's Hospital, Thane, IND
| | - Anil Hanumantharaju
- Neonatology, Pramukhswami Medical College, Bhaikaka University, Karamsad, IND
| | - Astha Amrit
- Neonatology, Mount Sinai Hospital, Toronto, CAN
| | - Somashekhar M Nimbalkar
- Central Research Services, Bhaikaka University, Karamsad, IND
- Pediatrics, Pramukhswami Medical College, Karamsad, IND
| | - Sanjay Patole
- Neonatology, King Edward Memorial Hospital, Perth, AUS
| |
Collapse
|
38
|
Darwesh MAK, Bakr W, Omar TEI, El-Kholy MA, Azzam NF. Unraveling the relative abundance of psychobiotic bacteria in children with Autism Spectrum Disorder. Sci Rep 2024; 14:24321. [PMID: 39414875 PMCID: PMC11484847 DOI: 10.1038/s41598-024-72962-3] [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: 05/28/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder characterized by social deficits. Accumulated evidence has shown a link between alterations in the composition of gut microbiota and both neurobehavioural and gastrointestinal symptoms in children with ASD which are related to the genera Lactobacillus and Bifidobacterium. These genera have been recently categorized as "psychobiotics". Moreover, this study aimed to compare the relative abundance of psychobiotics (L. plantarum, L. reuteri, and B. longum) to the total gut microbiome in typically developing (TD) children and those with ASD in order to correlate the distribution of psychobiotic with the severity and sensory impairments in autism. The ASD children were assessed using the Childhood Autism Rating Scale (CARS), while sensory impairments were evaluated using the Short Sensory Profile (SSP). Furthermore, the gut microbiome was analyzed using the quantitative real-time PCR. The study revealed a statistically significant increase in the relative abundance of L. reuteri and L. plantarum in the TD group in comparison to ASD children. Regarding the SSP total score of ASD children, a statistically significant negative correlation was found between both Lactobacillus and L. plantarum with the under-responsive subscale. For the Autism Treatment Evaluation Checklist (ATEC) score, B. longum and Lactobacillus showed a significant positive correlation with Health/Physical/Behaviour.
Collapse
Affiliation(s)
- Mennat-Allah K Darwesh
- Department of Microbiology. High Institute of Public Health, Alexandria University, Alexandria, Egypt.
| | - Wafaa Bakr
- Department of Microbiology. High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Tarek E I Omar
- Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohammed A El-Kholy
- Department of Microbiology and Biotechnology, Division of Clinical and Biological Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Alexandria, Egypt
| | - Nashwa Fawzy Azzam
- Department of Microbiology. High Institute of Public Health, Alexandria University, Alexandria, Egypt
| |
Collapse
|
39
|
Matar A, Damianos JA, Jencks KJ, Camilleri M. Intestinal Barrier Impairment, Preservation, and Repair: An Update. Nutrients 2024; 16:3494. [PMID: 39458489 PMCID: PMC11509958 DOI: 10.3390/nu16203494] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Our objective was to review published studies of the intestinal barrier and permeability, the deleterious effects of dietary components (particularly fat), the impact of altered intestinal permeability in disease models and human diseases, the role of the microbiome and epigenomics in control of barrier function, and the opportunities to restore normal barrier function with dietary interventions and products of the microbiota. METHODS We conducted a literature review including the following keywords alone or in combination: intestinal barrier, permeability, microbiome, epigenomics, diet, irritable bowel syndrome, inflammatory bowel disease, probiotics. RESULTS Intestinal permeability is modified by a diet including fat, which increases permeability, and nutrients such as fiber, glutamine, zinc, vitamin D, polyphenols, emulsifiers, and anthocyanins, which decrease permeability. There is significant interaction of the microbiome and barrier function, including the inflammatory of luminal/bacterial antigens, and anti-inflammatory effects of commensals or probiotics and their products, including short-chain fatty acids. Epigenomic modification of barrier functions are best illustrated by effects on junction proteins or inflammation. Detailed documentation of the protective effects of diet, probiotics, prebiotics, and microbiota is provided. CONCLUSION intestinal permeability is a critical factor in protection against gastrointestinal diseases and is impacted by nutrients that preserve or heal and repair the barrier and nurture anti-inflammatory effects.
Collapse
Affiliation(s)
| | | | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.); (J.A.D.)
| |
Collapse
|
40
|
Maniya H, Modasiya I, Chauhan M, Mori P, Kumar V. Developing Robust Probiotic Consortia: A Methodological Optimization Approach. Curr Microbiol 2024; 81:407. [PMID: 39404876 DOI: 10.1007/s00284-024-03933-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/01/2024] [Indexed: 11/21/2024]
Abstract
Developing effective probiotic consortia requires a comprehensive understanding of strain interactions. While traditional methods focus on direct interactions of the participating microbes, the role of microbial metabolites remains largely unexplored. Present study introduces a novel approach of evaluating the impact of strains as well as their secondary metabolites on compatibility during co-culture by assessing the antagonistic and synergistic attributes for multi-strain probiotic formulation. Assessment of antagonistic activity by spot method indicated suppressive nature of PIG1FD and PIG1IR on other strain's growth, hence not appropriate for consortia formulation. Findings of synergistic attribute demonstrated growth promoting role of cell supernatants from isolates PIG6IR and PIG5CI significantly, as it accelerated the entry of all other isolates into the log phase by 5-6 h and 0-2 h, respectively. By employing this methodology, we identified PIG5CI and PIG6IR (isolates identified as Bacillus spizizenii BAB 7915 and Bacillus subtilis BAB 7918 by 16S RNA sequencing method) as promising candidates for consortium formation due to their ability to enhance the growth of other strains through metabolite production. By attempting to elucidate the microbial interactions and metabolite-mediated effects, this research contributes to a more comprehensive understanding of probiotic consortia dynamics and offers valuable insights for future translational studies.
Collapse
Affiliation(s)
- Hina Maniya
- Postbiotic and Foodomics Lab, Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, 360020, India
| | - Ishita Modasiya
- Postbiotic and Foodomics Lab, Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, 360020, India
| | - Mehul Chauhan
- Postbiotic and Foodomics Lab, Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, 360020, India
| | - Priya Mori
- Postbiotic and Foodomics Lab, Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, 360020, India
| | - Vijay Kumar
- Postbiotic and Foodomics Lab, Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, 360020, India.
| |
Collapse
|
41
|
Aydin SS, Hatipoglu D. Probiotic strategies for mitigating heat stress effects on broiler chicken performance. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2024; 68:2153-2171. [PMID: 39320540 DOI: 10.1007/s00484-024-02779-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/17/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
The primary objective of this study was to evaluate the effects of liquid (Fructose-added lactic acid bacteria, F-LAB) and commercial (Commercial LAB, C-LAB) probiotics sourced from Rye-Grass Lactic Acid Bacteria (LAB) on broiler chickens experiencing heat stress (HS). The research involved 240 broiler chicks, divided into six groups: control, F-LAB, C-LAB (raised at 24 °C), HS, F-LAB/HS, and C-LAB/HS (exposed to 5-7 h of 34-36 °C daily). The study followed a randomized complete block design, with each group consisting of 40 chicks. F-LAB and HS/F-LAB groups received a natural probiotic added to their drinking water at a rate of 0.5 ml/L, while C-LAB and HS/C-LAB groups were supplemented with a commercial probiotic at the same dosage. Control and HS groups received no probiotic supplementation. The duration of the study was 42 days, with data collected on growth performance, feed intake, feed conversion ratio, and health parameters. Statistical analyses were performed using ANOVA, and significant differences between groups were determined using post hoc tests. The results revealed that without probiotic supplementation, heat stress led to a decrease in body weight gain, T3 levels, citrulline, and growth hormone levels, along with an increase in the feed conversion ratio, serum corticosterone, HSP70, ALT, AST, and leptin levels (p < 0.05 for all). Heat stress also adversely affected cecal microbiota, reducing lactic acid bacteria count (LABC) while increasing Escherichia coli and coliform bacteria (CBC) counts. However, in the groups receiving probiotic supplementation under heat stress (F-LAB/HS and C-LAB/HS), these effects were alleviated (p < 0.05 for all). Particularly noteworthy was the observation that broiler chickens supplemented with natural lactic acid bacteria (F-LAB) exhibited greater resilience to heat stress compared to those receiving the commercial probiotic, as evidenced by improvements in growth, liver function, hormonal balance, intestinal health, and cecal microbiome ecology (p < 0.05). These findings suggest that the supplementation of naturally sourced probiotics (F-LAB) may positively impact the intestinal health of broiler chickens exposed to heat stress, potentially supporting growth and health parameters.
Collapse
Affiliation(s)
- Sadik Serkan Aydin
- Department of Animal Nutrition and Nutritional Disease, Faculty of Veterinary Medicine, Harran University, Şanlıurfa, Türkiye
| | - Durmus Hatipoglu
- Department of Physiology, Faculty of Veterinary Medicine, Selcuk University, 42130, Konya, Türkiye.
| |
Collapse
|
42
|
Młynarska E, Budny E, Saar M, Wojtanowska E, Jankowska J, Marciszuk S, Mazur M, Rysz J, Franczyk B. Does the Composition of Gut Microbiota Affect Chronic Kidney Disease? Molecular Mechanisms Contributed to Decreasing Glomerular Filtration Rate. Int J Mol Sci 2024; 25:10429. [PMID: 39408756 PMCID: PMC11477184 DOI: 10.3390/ijms251910429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic kidney disease (CKD) is a very prevalent and insidious disease, particularly with initially poorly manifested symptoms that progressively culminate in the manifestation of an advanced stage of the condition. The gradual impairment of kidney function, particularly decreased filtration capacity, results in the retention of uremic toxins and affects numerous molecular mechanisms within the body. The dysbiotic intestinal microbiome plays a crucial role in the accumulation of protein-bound uremic toxins such as p-cresol (pC), indoxyl sulfate (IS), and p-cresyl sulfate (p-CS) through the ongoing fermentation process. The described phenomenon leads to an elevated level of oxidative stress and inflammation, subsequently resulting in tissue damage and complications, particularly an increase in cardiovascular risk, representing the predominant cause of mortality in chronic kidney disease (CKD). Therefore, exploring methods to reduce uremic toxins is currently a pivotal therapeutic strategy aimed at reducing the risk of organ damage in patients with chronic kidney disease (CKD). This review aims to summarize recent discoveries on modifying the composition of the intestinal microbiota through the introduction of special probiotic and synbiotic supplements for CKD therapy. The potential to connect the gut microbiota with CKD opens the possibility for further extensive research in this area, which could lead to the incorporation of synbiotics and probiotics into the fundamental treatment and prevention of CKD.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Emilian Budny
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Maciej Saar
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Ewa Wojtanowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Justyna Jankowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Szymon Marciszuk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Marcin Mazur
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (M.M.)
| |
Collapse
|
43
|
Liu CF, Chao WY, Shih TW, Lee CL, Pan TM. The Enhancement of Regulatory T Cell Maturation and Th1/Th2 Balance through FOXP3 Expression by Lactobacillus paracasei in an Ovalbumin-Induced Allergic Skin Animal Model. Curr Issues Mol Biol 2024; 46:10714-10730. [PMID: 39451516 PMCID: PMC11505879 DOI: 10.3390/cimb46100636] [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: 08/18/2024] [Revised: 09/14/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic allergic skin conditions, including atopic dermatitis (AD), are characterized by pruritus, erythema, xerosis, desquamation, and inflammation, significantly impacting quality of life. Long-term steroid use, while common in treatment, carries the risk of adverse effects. Previous studies have demonstrated the potential of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) in alleviating AD symptoms from a preventive perspective. This study, however, focuses on exploring NTU 101's therapeutic potential by investigating its effects on regulatory T cell (Treg) maturation and Th1/Th2 balance. The results revealed that NTU 101 administration effectively reduced serum IgE levels and inflammatory cell infiltration in the skin, leading to a significant improvement in both epidermal and dermal thickness in the AD model. Additionally, NTU 101 modulated the immune response by increasing the proportion of CD4+/IL-4+ (Th2) cells in the spleen and concurrently enhancing FOXP3 expression in CD4+/CD25+ cells, which is critical for Treg cell development. This immune modulation was further associated with a rebalancing of the Th1/Th2 ratio, achieved by increasing the proportion of CD4+/IFN-γ+ (Th1) cells. Moreover, NTU 101 influenced the proportion of CD4+IL-17+ (Th17) cells, thereby supporting neutrophil maturation and promoting allergen clearance, ultimately mitigating AD symptoms. These findings underscore the potential of NTU 101 not only in managing AD symptoms but also in modulating key immune pathways involved in the pathogenesis of the disease, offering a promising alternative or adjunct to conventional steroid therapies.
Collapse
Affiliation(s)
- Chin-Feng Liu
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung 950017, Taiwan;
| | - Wen-Yu Chao
- SunWay Biotech Co., Taipei 114063, Taiwan; (W.-Y.C.); (T.-W.S.)
| | - Tsung-Wei Shih
- SunWay Biotech Co., Taipei 114063, Taiwan; (W.-Y.C.); (T.-W.S.)
| | - Chun-Lin Lee
- Department of Life Science, National Taitung University, Taitung 950309, Taiwan
| | - Tzu-Ming Pan
- SunWay Biotech Co., Taipei 114063, Taiwan; (W.-Y.C.); (T.-W.S.)
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106216, Taiwan
| |
Collapse
|
44
|
Li P, Zhou H, Yang Y, Wu M, Zhao D, Wang L, Yi D, Hou Y. Dietary supplementation with N-acetylcysteine confers a protective effect on muscle and liver in lipopolysaccharide-challenged piglets. Front Nutr 2024; 11:1458912. [PMID: 39351494 PMCID: PMC11439690 DOI: 10.3389/fnut.2024.1458912] [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: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
N-acetylcysteine (NAC) is a well-established antioxidant that offers exciting opportunities for intestinal health in weaned piglets, while the effects of NAC on muscle and liver has not been fully characterized. Therefore, the present study was performed to investigate the effects of dietary supplementation with NAC on muscle and liver in weaned piglets challenged with lipopolysaccharide (LPS). Twenty-four piglets (24-day-old) were randomly assigned to three treatment groups, the piglets in the control (CTR) and LPS- challenged (LPS) groups were fed the basal diet and those in the LPS+ NAC group was fed the basal diet supplemented with 500 mg/kg NAC. The animal trial lasted for 21 days. At the end of the trial, piglets in the LPS and LPS+ NAC groups were injected intraperitoneally with LPS (100 μg/kg body weight) and piglets in the CTR group were administrated with an equal volume of normal saline. 3 h later, the blood was collected and tissue samples were obtained after 6 h of LPS or normal saline treatment. The results showed that the level of IL-1β, and the mRNA levels of C-X-C motif chemokine receptor 3 (CXCR3) and interferon-γ (IFN-γ) in the liver were up-regulated, and the mRNA levels of insulin-like growth factor 1 (IGF-1), total glutathione (T-GSH), and the ratio of total protein to DNA in the liver were decreased under LPS challenge (P < 0.05). At the same time, LPS increased the level of H2O2 and decreased the content of T-GSH and DNA in the longissimus dorsi and gastrocnemius muscles (P < 0.05). In addition, the percentage of monocytes and the level of epidermal growth factor (EGF) were down-regulated in the LPS treatment (P < 0.05). Interestingly, dietary NAC supplementation reversed the above changes induced by LPS (P < 0.05). Furthermore, NAC might alleviate the muscle and liver injury in LPS-challenged piglets by regulating the expression of genes related to the type I interferon signaling pathway, as well as hypoxia inducible factor 1 (HIF1) and nuclear factor erythroid-2 related factor 2 (Nrf-2). Our findings suggested that dietary supplementation with NAC could benefit the health of muscle and liver in LPS-challenged weaned piglets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yongqing Hou
- Engineering Research Center of Feed Protein Resources on Agricultural By-Products, Ministry of Education, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| |
Collapse
|
45
|
Gagliano C, Salvetat ML, Musa M, D'Esposito F, Rusciano D, Maniaci A, Pellegrini F, Scibilia G, Zeppieri M. Bacterial Insights: Unraveling the Ocular Microbiome in Glaucoma Pathogenesis. FRONT BIOSCI-LANDMRK 2024; 29:310. [PMID: 39206909 DOI: 10.31083/j.fbl2908310] [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: 04/11/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024]
Abstract
This review explores the connection between the ocular surface microbiome and glaucoma, highlighting its impact on disease progression. Beginning with an overview of global glaucoma significance, it emphasizes the importance of understanding the cellular characteristics and microbiology of the ocular microbiome. A search was conducted on the PubMed and Cochrane Library databases using the phrase "ocular microbiome glaucoma". 0 records were returned from the Cochrane Library while 21 were returned from PubMed. A total of 21 results were retrieved from 2017 to 2024. This comprised one opinion paper, four original research articles, and 16 reviews. This review covered the anatomy of the ocular surface, advanced analysis methods, and the ocular microbiome. It also delved into dysbiosis in glaucoma, addressing altered microbial communities and their potential role in disease progression. The intricate interplay between the ocular microbiome and the host's immune system is explored, emphasizing crosstalk and inflammatory responses. The review concludes by discussing therapeutic implications, including modulating ocular microbiota and potential future treatment strategies. Understanding the microbiome in healthy and glaucomatous eyes can help researchers and clinicians in innovative approaches to ocular health.
Collapse
Affiliation(s)
- Caterina Gagliano
- Department of Medicine and Surgery, University of Enna "Kore", 94100 Enna, Italy
- Eye Clinic, Catania University San Marco Hospital, 95121 Catania, Italy
| | - Maria Letizia Salvetat
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, 300238 Benin, Edo, Nigeria
| | - Fabiana D'Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, NW15QH London, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Napoli, Italy
| | | | - Antonino Maniaci
- Department of Medicine and Surgery, University of Enna "Kore", 94100 Enna, Italy
| | - Francesco Pellegrini
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Giuseppe Scibilia
- Department of Obstetrics and Gynecology, "Giovanni Paolo II" Hospital, 97100 Ragusa, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
46
|
Xiao N, Zhang X, Xi Y, Li Z, Wei Y, Shen J, Wang L, Qin D, Xie Z, Li Z. Study on the effects of intestinal flora on gouty arthritis. Front Cell Infect Microbiol 2024; 14:1341953. [PMID: 39176260 PMCID: PMC11339034 DOI: 10.3389/fcimb.2024.1341953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Gouty arthritis (GA), a metabolic and immunologic disease, primarily affects joints. Dysbiosis of intestinal flora is an important cause of GA. The metabolic disorders of intestinal flora leading to GA and immune disorders might play an important role in patients with hyperuricemia and established GA. However, the exact mechanisms, through which the dysbiosis of intestinal flora causes the development of GA, are not fully understood yet. Moreover, several therapies commonly used to treat GA might alter the intestinal flora, suggesting that modulation of the intestinal flora might help prevent or treat GA. Therefore, a better understanding of the changes in the intestinal flora of GA patients might facilitate the discovery of new diagnostic and therapeutic approaches. The current review article discusses the effects of intestinal flora dysbiosis on the pathogenesis of GA and the cross-regulatory effects between gut flora and drugs for treating GA. This article also highlights the modulatory effects of gut flora by traditional Chinese medicine (TCM) to lower uric acid levels and relieve joint pain as well as provides a summary and outlook, which might help guide future research efforts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dongdong Qin
- Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaofu Li
- Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
47
|
Li S, Liu Z, Zhang Q, Su D, Wang P, Li Y, Shi W, Zhang Q. The Antidiabetic Potential of Probiotics: A Review. Nutrients 2024; 16:2494. [PMID: 39125375 PMCID: PMC11313988 DOI: 10.3390/nu16152494] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetes has become one of the most prevalent global epidemics, significantly impacting both the economy and the health of individuals. Diabetes is associated with numerous complications, such as obesity; hyperglycemia; hypercholesterolemia; dyslipidemia; metabolic endotoxemia; intestinal barrier damage; insulin-secretion defects; increased oxidative stress; and low-grade, systemic, and chronic inflammation. Diabetes cannot be completely cured; therefore, current research has focused on developing various methods to control diabetes. A promising strategy is the use of probiotics for diabetes intervention. Probiotics are a class of live, non-toxic microorganisms that can colonize the human intestine and help improve the balance of intestinal microbiota. In this review, we summarize the current clinical studies on using probiotics to control diabetes in humans, along with mechanistic studies conducted in animal models. The primary mechanism by which probiotics regulate diabetes is improved intestinal barrier integrity, alleviated oxidative stress, enhanced immune response, increased short-chain fatty acid production, etc. Therefore, probiotic supplementation holds great potential for the prevention and management of diabetes.
Collapse
Affiliation(s)
- Shiming Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100193, China
| | - Zichao Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qi Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Dan Su
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14850, USA;
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Yixuan Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Wenbiao Shi
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| |
Collapse
|
48
|
Compare D, Sgamato C, Rocco A, Coccoli P, Ambrosio C, Nardone G. The Leaky Gut and Human Diseases: "Can't Fill the Cup if You Don't Plug the Holes First". Dig Dis 2024; 42:548-566. [PMID: 39047703 DOI: 10.1159/000540379] [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: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The gut barrier is a sophisticated and dynamic system that forms the frontline defense between the external environment and the body's internal milieu and includes various structural and functional components engaged not only in digestion and nutrient absorption but also in immune regulation and overall health maintenance. SUMMARY When one or more components of the intestinal barrier lose their structure and escape their function, this may result in a leaky gut. Mounting evidence emphasizes the crucial role of the gut microbiome in preserving the integrity of the gut barrier and provides insights into the pathophysiological implications of conditions related to leaky gut in humans. Assessment of intestinal permeability has evolved from invasive techniques to noninvasive biomarkers, but challenges remain in achieving consensus about the best testing methods and their accuracy. Research on the modulation of gut permeability is just starting, and although no medical guidelines for the treatment of leaky gut syndrome are available, several treatment strategies are under investigation with promising results. KEY MESSAGES This review discusses the composition of the intestinal barrier, the pathophysiology of the leaky gut and its implications on human health, the measurement of intestinal permeability, and the therapeutic strategies to restore gut barrier integrity.
Collapse
Affiliation(s)
- Debora Compare
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Costantino Sgamato
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Alba Rocco
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Pietro Coccoli
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Carmen Ambrosio
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| |
Collapse
|
49
|
Tullio V. Probiotic Yeasts: A Developing Reality? J Fungi (Basel) 2024; 10:489. [PMID: 39057374 PMCID: PMC11277836 DOI: 10.3390/jof10070489] [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: 05/23/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Yeasts are gaining increasing attention for their potential health benefits as probiotics in recent years. Researchers are actively searching for new yeast strains with probiotic properties (i.e, Debaryomyces hansenii; Kluyveromyces marxianus; Yarrowia lipolytica; Pichia hudriavzevii; and Torulaspora delbrueckii) from various sources, including traditional fermented foods, the human gut, and the environment. This exploration is expanding the pool of potential probiotic yeasts beyond the well-studied Saccharomyces boulardii. Research suggests that specific yeast strains possess properties that could be beneficial for managing conditions like inflammatory bowel disease, irritable bowel syndrome, skin disorders, and allergies. Additionally, probiotic yeasts may compete with pathogenic bacteria for adhesion sites and nutrients, thereby inhibiting their growth and colonization. They might also produce antimicrobial compounds that directly eliminate harmful bacteria. To achieve these goals, the approach that uses probiotics for human health is changing. Next-generation yeast probiotics are emerging as a powerful new approach in the field of live biotherapeutics. By using genetic engineering, scientists are able to equip these tools with specialized capabilities. However, most research on these probiotic yeasts is still in its early stages, and more clinical trials are needed to confirm their efficacy and safety for various health conditions. This review could provide a brief overview of the situation in this field.
Collapse
Affiliation(s)
- Vivian Tullio
- Department of Public Health and Pediatrics, University of Turin, via Santena 9; 10126 Turin, Italy
| |
Collapse
|
50
|
Liu H, Guo X, Jiang K, Shi B, Liu L, Hou R, Chen G, Farag MA, Yan N, Liu L. Dietary polyphenols regulate appetite mechanism via gut-brain axis and gut homeostasis. Food Chem 2024; 446:138739. [PMID: 38412807 DOI: 10.1016/j.foodchem.2024.138739] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/04/2024] [Accepted: 02/10/2024] [Indexed: 02/29/2024]
Abstract
Nowadays, due to the rise of fast-food consumption, the metabolic diseases are increasing as a result of high-sugar and high-fat diets. Therefore, there is an urgent need for natural, healthy and side-effect-free diets in daily life. Whole grain supplementation can enhance satiety and regulate energy metabolism, effects that have been attributed to polyphenol content. Dietary polyphenols interact with gut microbiota to produce intermediate metabolites that can regulate appetite while also enhancing prebiotic effects. This review considers how interactions between gut metabolites and dietary polyphenols might regulate appetite by acting on the gut-brain axis. In addition, further advances in the study of dietary polyphenols and gut microbial metabolites on energy metabolism and gut homeostasis are summarized. This review contributes to a better understanding of how dietary polyphenols regulate appetite via the gut-brain axis, thereby providing nutritional references for citizens' dietary preferences.
Collapse
Affiliation(s)
- Hongyan Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Xue Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Kexin Jiang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Boshan Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lingyi Liu
- Department of Food Science and Technology, University of Nebraska-Lincoln, NE, USA
| | - Ruyan Hou
- State Key Laboratory of Tea Plant Biology and Utilization, Key Laboratory of Food Nutrition and Safety, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei 230036, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, Key Laboratory of Food Nutrition and Safety, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei 230036, China
| | - Mohamed A Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Ning Yan
- Plant Functional Component Research Center, Tobacco Research Institute of Chinese Academy of Agricultural Sciences, No. 11 Forth Longitudinal Keyuan Rd, Laoshan District, Qingdao 266101, China
| | - Lianliang Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|