1
|
Han W, Xiong N, Huang L. Probiotics and nanoparticle-mediated nutrient delivery in the management of transfusion-supported diseases. Front Cell Infect Microbiol 2025; 15:1575798. [PMID: 40292219 PMCID: PMC12021914 DOI: 10.3389/fcimb.2025.1575798] [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: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Bone marrow is vital for hematopoiesis, producing blood cells essential for oxygen transport, immune defense, and clotting. However, disorders like leukemia, lymphoma, aplastic anemia, and myelodysplastic syndromes can severely disrupt its function, leading to life-threatening complications. Traditional treatments, including chemotherapy and stem cell transplants, have significantly improved patient outcomes but are often associated with severe side effects and limitations, necessitating the exploration of safer, more targeted therapeutic strategies. Nanotechnology has emerged as a promising approach for addressing these challenges, particularly in the delivery of nutraceuticals-bioactive compounds derived from food sources with potential therapeutic benefits. Despite their promise, nutraceuticals often face clinical limitations due to poor bioavailability, instability, and inefficient delivery to target sites. Nanoparticles offer a viable solution by enhancing the stability, absorption, and targeted transport of nutraceuticals to bone marrow while minimizing systemic side effects. This study explores a range of bone marrow disorders, conventional treatment modalities, and the potential of nanoparticles to enhance nutraceutical-based therapies. By improving targeted delivery and therapeutic efficacy, nanoparticles could revolutionize bone marrow disease management, providing patients with more effective and less invasive treatment options. These advancements represent a significant step toward safer and more efficient therapeutic approaches, ultimately improving patient prognosis and overall health.
Collapse
Affiliation(s)
- Wendao Han
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of
Medical Sciences, Meizhou, China
| | | | | |
Collapse
|
2
|
Han J, Ren Y, Zhang P, Fang C, Yang L, Zhou S, Ji Z. The effectiveness of treatment with probiotics in preventing necrotizing enterocolitis and related mortality: results from an umbrella meta-analysis on meta-analyses of randomized controlled trials. BMC Gastroenterol 2025; 25:245. [PMID: 40217146 PMCID: PMC11987312 DOI: 10.1186/s12876-025-03788-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
INTRODUCTION Probiotic supplementation has been proposed as a preventive measure for necrotizing enterocolitis (NEC) in preterm infants. This umbrella meta-analysis assesses the effects of probiotics, including single-strain and multi-strain formulations, on NEC and related mortality. METHODS A comprehensive search was conducted in PubMed, Scopus, ISI Web of Science, and Embase for studies up to August 2024. The AMSTAR2 tool assessed the quality of included studies. Meta-analysis studies were selected based on the PICOS framework, focusing on preterm neonates (< 37-week gestation), probiotic supplementation (single-strain or multi-strain), placebo or standard care comparison, and outcomes of NEC and mortality. Pooled relative risks (RR) and odds ratios (OR) with 95% confidence intervals (CI) were calculated using random-effects models. RESULTS Overall, 35 eligible studies were included into the study. Twenty-six and 32 probiotic intervention arms used single- and multi-strain probiotics, respectively. The findings revealed that probiotics decreased NEC significantly (ESRR: 0.51; 95% CI: 0.46, 0.55, p < 0.001, and ESOR: 0.59; 95%CI: 0.48, 0.72, P < 0.001), and mortality rate (ESRR: 0.72; 95% CI: 0.68, 0.76, P < 0.001, and ESOR: 0.77; 95%CI: 0.70, 0.84, p < 0.001). CONCLUSION The present review suggests that supplementation with probiotics reduced NEC and related mortality. Probiotic supplementation can be recognized as a NEC-preventing approach in preterm and very preterm infants, particularly Multi-strain probiotics.
Collapse
Affiliation(s)
- Jiaju Han
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Yufeng Ren
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China.
| | - Peini Zhang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Leilei Yang
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| | - Zhiqing Ji
- Department of Gastrointestinal Surgery, Taizhou Hospital, Wenzhou Medical University, No.105 Westgate Street, Linhai, 317000, China
| |
Collapse
|
3
|
Monaco M, Torazza C, Fedele E, Grilli M. The Impact of the Exposome on Alzheimer's Disease: The Influence of Nutrition. Int J Mol Sci 2025; 26:3015. [PMID: 40243652 PMCID: PMC11988514 DOI: 10.3390/ijms26073015] [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: 02/14/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, memory loss, and behavioural changes. While genetic predispositions and pathological processes have been the traditional focus, this review highlights the fundamental role of environmental factors, particularly nutrition, within the exposome framework in modulating the risk and progression of AD. The exposome, which includes the totality of environmental exposures in an individual's lifetime, provides a comprehensive approach to understanding the complex aetiology of AD. In this review, we explore the impact of dietary factors and cyclic nucleotide pathways (cAMP/cGMP) on AD, emphasizing the potential of dietary interventions as therapeutic strategies. We investigate key aspects of how nutrition affects the accumulation of β-amyloid, the aggregation of tau proteins, and neuroinflammation. We also examine the impact of specific nutrients on cognitive performance and the risk of AD. Additionally, we discuss the potential of nutraceuticals with anti-phosphodiesterase activity and the role of various animal models of AD (such as 5xFAD, 3xTg-AD, Tg2576, and APP/PS1 mice) in demonstrating the effects of dietary interventions on disease onset and progression.
Collapse
Affiliation(s)
- Martina Monaco
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Massimo Grilli
- Pharmacology and Toxicology Unit, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (C.T.); (E.F.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
4
|
Mattar L, Thalib HI, Alnuwaimi M, Alsaadi H, Allouji HA, Alyafei J, Alshowiman L, Alsobyani N, Hassan FES. Challenges of concurrent HIV infection in the course and management of Crohn's disease. J Med Life 2025; 18:171-178. [PMID: 40291934 PMCID: PMC12022739 DOI: 10.25122/jml-2024-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/12/2024] [Indexed: 04/30/2025] Open
Abstract
Crohn's disease (CD) is a chronic transmural bowel inflammation with a multifactorial etiology involving genetic predisposition and immune dysregulation in response to environmental triggers. In patients with human immunodeficiency virus (HIV), an already compromised immune system further complicates the progression and management of CD, creating unique therapeutic challenges. Probiotics have recently gained attention as a potential therapeutic option for CD, especially due to their role in modulating the gut microbiota. However, their effectiveness in patients with HIV, especially in enhancing and maintaining remissions, remains underexplored. This review aimed to examine how HIV infection influences the course of inflammatory bowel disease (IBD) and its impact on CD management strategies. A systematic literature search was conducted using Google Scholar, PubMed, Springer, and Web of Science to identify studies on patients with HIV and CD. HIV infection significantly alters the progression and management of CD due to its impact on the immune system. The immunosuppressed state of patients with HIV can complicate both the diagnosis and treatment of CD, often requiring adjustments in therapeutic approaches, necessitating a careful, tailored approach.
Collapse
Key Words
- AAD, Antibiotic-Associated Diarrhea
- AIDS, Acquired Immunodeficiency Syndrome
- AIEC, Adherent-Invasive Escherichia Coli
- APC, Antigen-Presenting Cells
- ART, Antiretroviral Therapy
- CARD15, Caspase Recruitment Domain–Containing Protein 15
- CARD9, Caspase Recruitment Domain–Containing Protein 9
- CAZymes, Carbohydrate-Active Enzymes
- CCL4, C-C Motif Chemokine Ligand 4
- CCR5, C-C Chemokine Receptor Type 5
- CD, Cluster Of Differentiation
- CD, Crohn’s Disease
- CRC, Colorectal Cancer
- CXCR4, C-X-C Chemokine Receptor Type 4
- Crohn’s disease
- DC, Dendritic Cells
- DC-SIGN, Dendritic Cell–Specific Intercellular Adhesion Molecule-3–Grabbing Non-Integrin
- ERS, Endoplasmic Reticulum Stress
- FMT, Fecal Microbiota Transplantation
- FVT, Fecal Virome Transplantation
- GIT, Gastrointestinal Tract
- HIV
- HIV, Human Immunodeficiency Virus
- IBD, Inflammatory Bowel Disease
- IFABP, Intestinal Fatty Acid–Binding Protein
- IL, Interleukin
- ILCs, Innate Lymphoid Cells
- MALT, Mucosa-Associated Lymphoid Tissue
- MAMP, Microbe-Associated Molecular Pattern
- NF-κB, Nuclear Factor Kappa B
- NK, Natural Killer Cells
- NOD2, Nucleotide-Binding Oligomerization Domain–Containing Protein 2
- NOS, Nitric Oxide Synthase
- PPAR-γ, Peroxisome Proliferator-Activated Receptor Gamma
- PRR, Pattern Recognition Receptor
- SCFA, Short-Chain Fatty Acids
- SLE, Systemic Lupus Erythematosus
- TGF-β, Transforming Growth Factor–β
- TLR, Toll-Like Receptor
- TNF-α, Tumor Necrosis Factor–α
- Th17, T Helper 17 Cells
- UC, Ulcerative Colitis
- gut microbiota
- pDC, Plasmacytoid Dendritic Cells
- probiotics
- sCD14, Soluble CD14
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fatma El Sayed Hassan
- Medical Physiology Department, Kasr Alainy Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
Reiriz M, Beltrán-Velasco AI, Echeverry-Alzate V, Martínez-Miguel E, Gómez-Senent S, Uceda S, Clemente-Suárez VJ. Bifidobacterium infantis and Bifidobacterium breve Improve Symptomatology and Neuronal Damage in Neurodegenerative Disease: A Systematic Review. Nutrients 2025; 17:391. [PMID: 39940249 PMCID: PMC11820578 DOI: 10.3390/nu17030391] [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: 11/25/2024] [Revised: 01/18/2025] [Accepted: 01/19/2025] [Indexed: 02/14/2025] Open
Abstract
Background/Objectives: This systematic review focused on collecting the most significant findings on the impact of the administration of Bifidobacterium infantis (or Bifidobacterium longum subps. infantis) and Bifidobacterium breve, alone, in conjunction, or in combination with other strains, in the treatment of neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases are characterized by the progressive degeneration of neurons, resulting in a broad spectrum of clinical manifestations. AD is typified by a progressive decline in cognitive abilities, while PD is marked by motor symptoms associated with the loss of dopamine (DA). Methods: Five different databases, ScienceDirect, Scopus, Wiley, PubMed, and Web of Science (WoS), were reviewed and the studies were screened for inclusion by the following criteria: (i) studies that specifically evaluated the use of Bifidobacterium infantis, Bifidobacterium longum subsp. infantis, or Bifidobacterium breve as a therapeutic intervention, either in human or animal models, in the context of neurodegenerative diseases; (ii) the studies were required to address one or more of the pathologies examined in this article, and the pathologies included, but were not limited to, neurodegeneration, Alzheimer's disease, Parkinson's disease, and oxidative stress; (iii) the full text was accessible online; and (iv) the article was written in English. Results: The data suggest that these probiotics have neuroprotective effects that may delay disease progression. Conclusions: This study provides updated insights into the use of these Bifidobacterium strains in neurodegenerative diseases like AD and PD, with the main limitation being the limited number of clinical trials available.
Collapse
Affiliation(s)
- Manuel Reiriz
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Ana Isabel Beltrán-Velasco
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Víctor Echeverry-Alzate
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Esther Martínez-Miguel
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Silvia Gómez-Senent
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Sara Uceda
- NBC Group, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain; (M.R.); (A.I.B.-V.); (V.E.-A.); (E.M.-M.); (S.G.-S.)
| | - Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
6
|
Jiang Q, Zhu X, Sun L, Xie C, Wang X, Ma L, Yan X. Akkermansia muciniphila Promotes SIgA Production and Alters the Reactivity Toward Commensal Bacteria in Early-Weaned Piglets. J Nutr 2025; 155:52-65. [PMID: 39528052 DOI: 10.1016/j.tjnut.2024.11.002] [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: 07/09/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Secretory IgA (SIgA) is the first line of defense in protecting the intestinal epithelium against pathogenic bacteria, regulating gut microbiota composition, and maintaining intestinal homeostasis. Early weaning strategies may disrupt SIgA levels in piglet intestines, causing a decline in immune response and early weaning stress. However, the specific microbial mechanisms modulating SIgA in early-weaned piglets are not well understood. OBJECTIVES We hypothesized that Akkermansia muciniphila increases intestinal SIgA production in the early-weaned piglets. METHODS Fecal SIgA levels, SIgA-coated bacteria abundance, and fecal metagenomes were compared between 6 Huanjiang miniature (HM) and 6 Duroc×Landrace×Yorkshire (DLY) early-weaned piglets to identify bacterial species involved in SIgA modulation. Four bacterial species were investigated using 5 groups (Control, A. muciniphila, L. amylovorus, L. crispatus, and L. acidophilus) of male specific pathogen-free C57BL/6J mice, weaned 3 wk postbirth (n = 8/group). Subsequently, 10-d-old Landrace×Yorkshire (LY) piglets were randomly assigned to 3 groups (Control, 109A. muciniphila, and 108A. muciniphila) (n = 10/group) to evaluate the effect of orally administered A. muciniphila on intestinal SIgA production and microbial composition. RESULTS HM early-weaned piglets showed significantly higher SIgA levels [7.59 μg/mg, 95% confidence interval (CI): 3.2, 12, P = 0.002] and SIgA-coated bacteria abundance (8.64%, 95% CI: 3.2, 14, P = 0.014) than DLY piglets. In the mouse model, the administration of A. muciniphila significantly increased SIgA levels (3.50 μg/mg, 95% CI: 0.59, 6.4, P = 0.018), SIgA-coated bacteria abundance (9.06%, 95% CI: 4, 14, P = 0.018), and IgA+ plasma cell counts (6.1%, 95% CI: 4.3, 8, P = 0.005). In the pig experiments, the oral administration of A. muciniphila to LY piglets significantly enhanced intestinal SIgA concentrations (4.22 μg/mg, 95% CI: 0.37, 8.5, P = 0.034) and altered the SIgA-coated bacterial landscape. CONCLUSIONS Early intervention with A. muciniphila in nursing piglets can increases intestinal SIgA production and alter the reactivity toward commensal bacteria upon early weaning.
Collapse
Affiliation(s)
- Qin Jiang
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China; Yazhouwan National Laboratory (YNL), Sanya, China
| | - Xiaoyan Zhu
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lingling Sun
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chunlin Xie
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xinkai Wang
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Libao Ma
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xianghua Yan
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Nami Y, Barghi A, Shahgolzari M, Salehian M, Haghshenas B. Mechanism of Action and Beneficial Effects of Probiotics in Amateur and Professional Athletes. Food Sci Nutr 2025; 13:e4658. [PMID: 39803224 PMCID: PMC11717059 DOI: 10.1002/fsn3.4658] [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: 05/25/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, provide health benefits to the host. According to the International Society of Sports Nutrition (ISSN), probiotic supplementation can optimize the health, performance, and recovery of athletes at all stages of their careers. Recent research suggests that probiotics can improve immune system functions, reduce gastrointestinal distress, and increase gut permeability in athletes. Additionally, probiotics may provide athletes with secondary health benefits that could positively affect athletic performance through enhanced recovery from fatigue, improved immune function, and maintenance of healthy gastrointestinal tract function. The integration of some probiotic strains into athletes' diets and the consumption of multi-strain compounds may lead to an improvement in performance and can positively affect performance-related aspects such as fatigue, muscle pain, body composition, and cardiorespiratory fitness. In summary, probiotics can be beneficial for athletes at all stages of their careers, from amateur to professional. This paper reviews the progress of research on the role of probiotic supplementation in improving energy metabolism and immune system functions, reducing gastrointestinal distress, and enhancing recovery from fatigue in athletes at different levels.
Collapse
Affiliation(s)
- Yousef Nami
- Department of Food Biotechnology, Branch for Northwest and West RegionAgricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO)TabrizIran
| | - Anahita Barghi
- Institute of Agricultural Life ScienceDong‐A UniversityBusanSouth Korea
| | - Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Melika Salehian
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Babak Haghshenas
- Regenerative Medicine Research Center (RMRC), Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
8
|
Dabous A, Stellavato A, Cimini D, Vassallo V, D'Agostino M, Schiraldi C. A probiotic multi-strain mixture combined with hydroxyectoine improves intestinal barrier function by alleviating inflammation in lipopolysaccharide stimulated differentiated Caco-2 cells. Food Funct 2024; 15:11578-11589. [PMID: 39508078 DOI: 10.1039/d4fo03130j] [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: 11/08/2024]
Abstract
Many studies have highlighted the role of probiotics in re-establishing the gut microbiota balance and preventing intestinal barrier dysfunction. In fact, they can also contribute to the upregulation of anti-inflammatory genes and the downregulation of pro-inflammatory genes, which are known to contribute to the development of the inflammatory bowel disease (IBD) syndrome. The present study aims to investigate the effect of the compatible solute hydroxyectoine (HOE), to be used as a cryopreservant but also for its intrinsic biological properties, to obtain a new formula containing three probiotic strains (Limosilactobacillus fermentum (L. fermentum), Levilactobacillus brevis SP-48 (L. brevis), and Bifidobacterium lactis HN019 (B. lactis)), and evaluate the latter for its ability to prevent lipopolysaccharide (LPS)-induced inflammation in an in vitro bi-dimensional model of the intestinal barrier using a Caco-2 cell monolayer. The mRNA expression levels of the inflammatory cytokines (IL-6, IL-1β, and TNF-α) were analyzed by real-time PCR. Changes in the modulation of (TLR-4 and NF-κB) proteins were assessed by western blotting, and the effect of the HOE/PRO formula on the intestinal epithelial barrier function was also assessed using an immunofluorescence microscope for the tight junction protein zonula occludens-1 (ZO-1). This study found that this novel probiotic formulation containing HOE is capable of decreasing LPS-induced cytokines, as confirmed by the results of RT-PCR and ELISA and preserving the integrity of tight junctions as demonstrated by the relevant expression of ZO-1. HOE/PRO was shown to be effective in reducing the expression of TLR-4 and NF-κB. The latter plays a key role as an inflammation modulator as shown through experiments run with the THP-1/NF-κB reporter gene. Collectively, our data indicate that the HOE/PRO formula is a good candidate for potential preventive and/or therapeutic implementation in IBD.
Collapse
Affiliation(s)
- Azza Dabous
- Department of Experimental Medicine, Section of Biotechnology, Molecular Biology and Medical Histology, University of Campania "Luigi Vanvitelli", Naples, Italy.
- Department of Nutrition and Food Technology, An-Najah National University, P.O. Box 7, Nablus, Palestine
| | - Antonietta Stellavato
- Department of Experimental Medicine, Section of Biotechnology, Molecular Biology and Medical Histology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Donatella Cimini
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Valentina Vassallo
- Department of Experimental Medicine, Section of Biotechnology, Molecular Biology and Medical Histology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Maria D'Agostino
- Department of Experimental Medicine, Section of Biotechnology, Molecular Biology and Medical Histology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Chiara Schiraldi
- Department of Experimental Medicine, Section of Biotechnology, Molecular Biology and Medical Histology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
9
|
Ambe NF, Bobga TP, Toukam Tatsinkou LL, Sotoing Taiwe G, Fossi BT. Ameliorative effects of probiotic Limosilactobacillus fermentum and Enterococcus lactis isolated from cameroonian traditionally processed milk and palm wine against chronic constriction injury induced neuropathic pain in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118560. [PMID: 39004193 DOI: 10.1016/j.jep.2024.118560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fermented milk and palm wine are regularly used by several ethnic groups in Cameroon in traditional treatment rituals for infections, inflammatory, cardiovascular disorders, and even metabolic diseases such as diabetes, hypercholesterolemia etc. Reports from many studies have demonstrated that fermented milk and palm wine are potential sources of probiotic bacteria. However, the capacity of probiotics isolated from these natural sources to alleviate neuropathic pain has not been experimentally tested. AIM OF THE STUDY This study aimed at investigating the ameliorative potential of lactic acid bacteria isolated from palm wine and traditional fermented cow milk on the chronic constriction injury (CCI) induced neuropathic pain in mice. MATERIALS AND METHODS Pour plating technique on De Man Rogasa (MRS) agar was utilised for isolation of lactic acid bacteria from fermented cow milk and palm wine, and identified using the 16S r RNA gene sequencing. Neuropathic pain was induced by chronic constriction injury of the sciatic nerve. These bacteria were orally administered at different concentrations to Balb/c mice by gavage for 14 consecutive days. Cold allodynia, mechanical hyperalgesia and exploratory behaviour were evaluated on day 0, 7th and 14th respectively. The total level of calcium, oxidative stress markers and myeloperoxidase were also quantified in the sciatic nerve homogenate. Cyclooxygenase-2(COX-2) and cytokine profile were determined from serum. RESULTS Lactic acid bacteria were isolated from fermented cow milk and palm wine and two isolates were chosen according to their probiotic potentials and identified as strain of Limolactobacillus fermentum and Enterococcus lactis. Their 16 S rRNA gene sequences were deposited in NCBI genbank with accession number of OP896078 and OR619545, respectively. Pretreatment with Limosilactobacillus fermentum and Enterococcus lactis significantly alleviated mechanical hyperalgesia and cold allodynia with similar effect to the reference drug, morphine. These two isolates ameliorated CCI induced neuropathic pain by increasing antioxida776nts (GSH, CAT and SOD, P < 0.01) and decreasing pro-oxidants (MDA and NO, P < 0.01). Also, they inhibited the release of proinflammatory cytokines (IL-1β, TNF-α, IFN-γ, and IL-6; P < 0.01) and IL-10 level was significantly (P < 0.01) increased when compared to the negative control. Treatment with these bacteria significantly dropped the level of total calcium (P < 0.01), COX-2 (P < 0.01) and MPO (P < 0.01) when compared with the negative control. CONCLUSION The neuroprotective potentials of these selected lactic acid bacteria against CCI induced neuropathic pain may be attributed to their anti-oxidant, anti-inflammatory properties and reduced calcium deposition in sciatic nerve.
Collapse
Affiliation(s)
- Ngwa Fabrice Ambe
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Cameroon; Department of Medical Laboratory Sciences, Faculty Health Sciences, University of Buea, Cameroon
| | - Tanyi Pride Bobga
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, Cameroon; Department of Medical Laboratory Sciences, Faculty Health Sciences, University of Buea, Cameroon
| | | | - Germain Sotoing Taiwe
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Cameroon.
| | | |
Collapse
|
10
|
Horvath A, Habisch H, Prietl B, Pfeifer V, Balazs I, Kovacs G, Foris V, John N, Kleinschek D, Feldbacher N, Grønbæk H, Møller HJ, Žukauskaitė K, Madl T, Stadlbauer V. Alteration of the Gut-Lung Axis After Severe COVID-19 Infection and Modulation Through Probiotics: A Randomized, Controlled Pilot Study. Nutrients 2024; 16:3840. [PMID: 39599626 PMCID: PMC11597208 DOI: 10.3390/nu16223840] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The gut-lung axis could be a potential therapeutic target for improving post-acute COVID-19 symptoms, and probiotics have been proposed as possible modulators. AIM We conducted a pilot study to understand alterations in the gut-lung axis and to explore the effects of a probiotic in post-acute COVID-19 disease. METHODS We included patients after severe COVID-19 disease (sCOV, n = 21) in a randomized, placebo-controlled trial to test the effect of a probiotic (Pro-Vi 5, Institute Allergosan, Graz, Austria) in a six-month intervention and used patients after mild disease (mCOV, n = 10) as controls, to compare the intestinal microbiome, metabolome, and patient-reported outcomes and biomarkers along the gut-lung axis at baseline and throughout probiotic intervention. RESULTS Compared to mCOV patients, sCOV patients showed lower microbial richness, which was significantly improved by probiotic intervention. A reorganization of Ruminococcaceae and Lachnospiraceae taxa was observed in sCOV patients but remained unaffected by the intervention. Serum metabolome showed a dysregulation of lipoproteins in accordance with higher BMI and comorbidities in sCOV patients. HDL and LDL fractions/components were temporarily decreased in the probiotic group. Stool metabolome was altered at baseline in sCOV patients and an increase in L-DOPA after 3 months and butyrate after 6 months of intervention could be observed. Probiotics partially improved reduced quality of life and modulated altered immune responses in sCOV patients. Increased intestinal permeability at baseline remained unaffected. CONCLUSION The study provides evidence of long-term alterations of the gut-lung axis after severe COVID-19 infection and suggests that probiotics can modulate the biomarkers of the gut-lung axis.
Collapse
Affiliation(s)
- Angela Horvath
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Hansjörg Habisch
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Barbara Prietl
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Verena Pfeifer
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division of Endocrinology and Diabetes, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Irina Balazs
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Gabor Kovacs
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Nikolaus John
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria; (G.K.); (V.F.); (N.J.)
| | - Daniela Kleinschek
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
| | - Nicole Feldbacher
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
| | - Henning Grønbæk
- Departments of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Kristina Žukauskaitė
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
- Institute of Biosciences, Life Sciences Center, Vilnius University, 01513 Vilnius, Lithuania
| | - Tobias Madl
- Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (T.M.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Vanessa Stadlbauer
- Center for Biomarker Research in Medicine (CBmed), Division of Translational Precision Medicine, Division of Precision Medicine Technologies, 8010 Graz, Austria; (A.H.); (B.P.); (V.P.); (I.B.); (N.F.)
- Division for Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria;
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
11
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
12
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
13
|
Yuan M, Chang L, Gao P, Li J, Lu X, Hua M, Li X, Liu X, Lan Y. Synbiotics containing sea buckthorn polysaccharides ameliorate DSS-induced colitis in mice via regulating Th17/Treg homeostasis through intestinal microbiota and their production of BA metabolites and SCFAs. Int J Biol Macromol 2024; 276:133794. [PMID: 38992530 DOI: 10.1016/j.ijbiomac.2024.133794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic condition whose incidence has been rising globally. Synbiotic (SYN) is an effective means of preventing IBD. This study investigated the preventive effects and potential biological mechanisms of SYN (Bifidobacterium longum, Lactobacillus acidophilus, and sea buckthorn polysaccharides) on DSS-induced colitis in mice. The results indicated that dietary supplementation with SYN has a significant improvement effect on DSS mice. SYN ameliorated disease activity index (DAI), colon length, and intestinal barrier permeability in mice. In addition, RT-qPCR results indicated that after SYN intervention, the expression levels of pro-inflammatory factors (IL-6, IL-1β, TNF-α, and IL-17F) and transcription factor RORγt secreted by Th17 cells were significantly reduced, and the expression levels of anti-inflammatory factors (IL-10 and TGF-β) and transcription factor Foxp3 secreted by Treg cells were robustly increased. 16S rDNA sequencing analysis revealed that key intestinal microbiota related to Th17/Treg balance (Ligilactobacillus, Lactobacillus, Bacteroides, and Akkermansia) was significantly enriched. At the same time, a significant increase in microbial metabolites SCFAs and BAs was observed. We speculate that SYN may regulate the Th17/Treg balance by restructuring the structure and composition of the intestinal microbiota, thereby mitigating DSS-induced colitis.
Collapse
Affiliation(s)
- Mingyou Yuan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Lili Chang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Pan Gao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jing Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xinyuan Lu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Mingfang Hua
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiulian Li
- School of Pharmacy, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Ying Lan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|
14
|
Patibandla S, Bhatt N, Lief S, Beauti SM, Ansari AZ. Gut Microbiota Modulation in the Management of Chronic Obstructive Pulmonary Disease: A Literature Review. Cureus 2024; 16:e66875. [PMID: 39280360 PMCID: PMC11398707 DOI: 10.7759/cureus.66875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) represents a significant global health burden, characterized by progressive airflow limitation and exacerbations that significantly impact patient morbidity and mortality. Recent research has investigated the interplay between the gut and the lungs, known as the gut-lung axis, highlighting the role of the gut microbiome in COPD pathogenesis. Dysbiosis, characterized by microbial imbalance, has implications for COPD, influencing disease progression and susceptibility to exacerbations. This comprehensive review integrates current scientific literature on gut microbiota modulation as a therapeutic avenue for COPD management. Through a thorough discussion of studies investigating probiotics, prebiotics, synbiotics, antibiotics, dietary fiber, and fecal microbiota transplantation, this review summarizes the influence of these interventions on COPD via the gut-lung axis through the modulation of systemic inflammation, mucosal immunity, and metabolic processes. The interventions highlighted here show potential in preventing COPD exacerbations, preserving lung function, and improving patient quality of life. By compiling the latest scientific evidence, this review provides a comprehensive framework for physicians and researchers to deduce the effectiveness of gut microbiome modulation as an adjunctive therapeutic strategy in COPD management.
Collapse
Affiliation(s)
- Srihita Patibandla
- Department of Internal Medicine, Trinity Health Grand Rapids, Grand Rapids, USA
| | - Nilay Bhatt
- Department of Internal Medicine, Hospital Corporation of America (HCA) Houston Healthcare Clear Lake, Webster, USA
| | - Sean Lief
- Department of Internal Medicine, William Carey University College of Osteopathic Medicine, Hattiesburg, USA
| | - Samer M Beauti
- Department of Internal Medicine, William Carey University College of Osteopathic Medicine, Hattiesburg, USA
| | - Ali Z Ansari
- Department of Pathology, William Carey University College of Osteopathic Medicine, Hattiesburg, USA
| |
Collapse
|
15
|
Yu T, Wu B, Zhang D, Deng G, Luo Y, Tang N, Shi Q, Hu F, Zhang G. A novel Bacillus aerolatus CX253 attenuates inflammation induced by Streptococcus pneumoniae in childhood and pregnant rats by regulating gut microbiome. Cell Mol Life Sci 2024; 81:319. [PMID: 39078497 PMCID: PMC11335247 DOI: 10.1007/s00018-024-05232-0] [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: 11/27/2023] [Revised: 01/20/2024] [Accepted: 04/05/2024] [Indexed: 07/31/2024]
Abstract
Streptococcus pneumoniae (Spn) is the predominant pathogen responsible for community-acquired pneumonia (CAP) in children under five years old, and it can induce over 17% of pregnant women. However, no more effective measures exist to prevent infection induced by Spn in these two special populations. The beneficial microbes can antagonize Spn and provide new targets for preventing pneumococcal infections. This study used 16S rRNA gene sequencing and targeted metabolomics to evaluate the role of the Bacillus aerolatus CX253 (CX253) in alleviating Spn infection. Additionally, the colonization of CX253 was observed in nose, trachea, and lung by using confocal laser scanning microscopy and fluorescent labeling techniques. Compared with the model group, the expression level of interleukin-1β was dropped 1.81-fold and 2.22-fold, and interleukin-6 was decreased 2.39-fold and 1.84-fold. The express of tumor necrosis factor-α was down 2.30-fold and 3.84-fold in prevention group of childhood and pregnant rats, respectively. The 16S rRNA sequencing results showed that CX253 administration alone significantly increased the abundance of Lactobacillus, Limosilactobacillus, and Prevotella in the gut of childhood and pregnant rats. Furthermore, the CX253 increased propionate in the gut of childhood rats and increased propionate and butyrate in the gut of pregnant rats to inhibit pulmonary inflammation. In summary, CX253 attenuated Spn-induced inflammation by regulating the gut microbiota and SCFAs. The research provides valuable information for the prevention of pneumonia.
Collapse
Affiliation(s)
- Ting Yu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Biru Wu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Dimei Zhang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Guanhua Deng
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, 1Tianqiang St., Huangpu West Ave, Guangzhou, 510620, Guangdong, China.
| | - Yi Luo
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ningqianzi Tang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Qiankun Shi
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Fang Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China.
| | - Guoxia Zhang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
16
|
Ermann Lundberg L, Pallabi Mishra P, Liu P, Forsberg MM, Sverremark-Ekström E, Grompone G, Håkansson S, Linninge C, Roos S. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024; 90:e0024724. [PMID: 38888338 PMCID: PMC11267924 DOI: 10.1128/aem.00247-24] [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: 02/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Collapse
Affiliation(s)
- Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| | - Punya Pallabi Mishra
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Sebastian Håkansson
- BioGaia, Stockholm, Sweden
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | - Caroline Linninge
- BioGaia, Stockholm, Sweden
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| |
Collapse
|
17
|
Bosselaar S, Dhelin L, Dautel E, Titecat M, Duthoy S, Stelmaszczyk M, Delory N, De Sousa Violante M, Machuron F, Ait-Abderrahim H, Desreumaux P, Foligné B, Monnet C. Taxonomic and phenotypic analysis of bifidobacteria isolated from IBD patients as potential probiotic strains. BMC Microbiol 2024; 24:233. [PMID: 38951788 PMCID: PMC11218132 DOI: 10.1186/s12866-024-03368-4] [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: 02/14/2024] [Accepted: 06/12/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Inflammatory Bowel Diseases (IBD) are a major public health issue with unclear aetiology. Changes in the composition and functionality of the intestinal microbiota are associated with these pathologies, including the depletion of strict anaerobes such as Feacalibacterium prausnitzii. Less evidence is observed for depletion in other anaerobes, among which bifidobacteria. This study characterized the taxonomic and functional diversity of bifidobacteria isolated from the human intestinal microbiota in active and non-active IBD patients by a culturomics approach and evaluated if these bifidobacteria might be used as probiotics for gut health. RESULTS A total of 341 bifidobacteria were isolated from the intestinal microbiota of IBD patients (52 Crohn's disease and 26 ulcerative colitis patients), with a high proportion of Bifidobacterium dentium strains (28% of isolated bifidobacteria). In ulcerative colitis, the major species identified was B. dentium (39% of isolated bifidobacteria), in active and non-active ulcerative colitis. In Crohn's disease, B. adolescentis was the major species isolated from non-active patients (40%), while similar amounts of B. dentium and B. adolescentis were found in active Crohn's disease patients. The relative abundance of B. dentium was increased with age, both in Crohn's disease and ulcerative colitis and active and non-active IBD patients. Antibacterial capacities of bifidobacteria isolated from non-active ulcerative colitis against Escherichia coli LF82 and Salmonella enterica ATCC 14028 were observed more often compared to strains isolated from active ulcerative colitis. Finally, B. longum were retained as strains with the highest probiotic potential as they were the major strains presenting exopolysaccharide synthesis, antibacterial activity, and anti-inflammatory capacities. Antimicrobial activity and EPS synthesis were further correlated to the presence of antimicrobial and EPS gene clusters by in silico analysis. CONCLUSIONS Different bifidobacterial taxonomic profiles were identified in the microbiota of IBD patients. The most abundant species were B. dentium, mainly associated to the microbiota of ulcerative colitis patients and B. adolescentis, in the intestinal microbiota of Crohn's disease patients. Additionally, the relative abundance of B. dentium significantly increased with age. Furthermore, this study evidenced that bifidobacteria with probiotic potential (antipathogenic activity, exopolysaccharide production and anti-inflammatory activity), especially B. longum strains, can be isolated from the intestinal microbiota of both active and non-active Crohn's disease and ulcerative colitis patients.
Collapse
Affiliation(s)
- Sabine Bosselaar
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France.
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France.
| | - Lucile Dhelin
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Ellena Dautel
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Marie Titecat
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Stéphanie Duthoy
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Marie Stelmaszczyk
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Nathan Delory
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Madeleine De Sousa Violante
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - François Machuron
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Hassina Ait-Abderrahim
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| | - Pierre Desreumaux
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
- Department of Hepato-Gastroenterology, Lille University Hospital, 59037, Lille, France
| | - Benoit Foligné
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Céline Monnet
- Lesaffre International - Lesaffre Institute of Science and Technology, 101 Rue de Menin, 59706, Marcq-en-Barœul, France
| |
Collapse
|
18
|
Roozbahani F, Ahanjan M, Moshiri M, Abediankenari S, Goli HR, Kakavan M, Gholami M. Characterization of Antimicrobial Activities of Bifidobacterium lactis BB-12 and Their Inhibitory Effect Against Some Foodborne Pathogens. Foodborne Pathog Dis 2024; 21:370-377. [PMID: 38578010 DOI: 10.1089/fpd.2023.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
Bifidobacterium animalis subsp. lactis BB-12, a probiotic, has shown potential to promote health benefits and control pathogens. This study aimed to investigate the effectiveness of BB-12 and its cell-free supernatant (CFS) in inhibiting the growth of Listeria monocytogenes and Salmonella enterica serovar Typhimurium. To assess the antimicrobial activity of BB-12, agar well diffusion, disk diffusion, and minimum inhibitory concentration (MIC) tests were conducted. The bicinchoninic acid (BCA) assay was performed to measure the protein concentration in CFS. The study's results indicated that the BB-12 strain inhibited the pathogens' growth. The disk diffusion test using BB-12 showed inhibitory results ranging from 11 to 14 mm for both bacteria. The agar well diffusion test reported the zone of inhibition ranging from 11.6 to 16 mm for both bacteria. The MIC test was conducted as a confirmatory test, which demonstrated the highest inhibitory zone using 2 McFarland (6 × 108 CFU/mL) concentrations of probiotics on L. monocytogenes (44.98%) and S. Typhimurium (66.41%). The disk diffusion test revealed that the probiotic CFS had a significant inhibitory impact on S. Typhimurium with a 16.6 mm zone of inhibition. The BCA test findings indicated that the 24- and 48-h CFSs exhibited inhibitory properties against infections. Notably, the 24-h CFS, including a protein level of 78.47 μg/mL, demonstrated a more pronounced inhibitory impact on both pathogens. The findings highlight that utilizing the BB-12 strain and its CFS can serve as a viable approach to battle infections, enhancing food safety and public health.
Collapse
Affiliation(s)
- Fatemeh Roozbahani
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ahanjan
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mona Moshiri
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maedeh Kakavan
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehrdad Gholami
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
19
|
Cao Z, Ling X, Haseeb A, Sun P, Zhang H, Yin W, Fan K, Yang H, Zhang Z, Zhong J, Sun Y, Sun N, Li H. Analysis of the anti-PCV2 mechanism of Lactobacillus acidophilus based on non-target metabolomics and high-throughput molecular docking. Front Microbiol 2024; 15:1416235. [PMID: 38860222 PMCID: PMC11163031 DOI: 10.3389/fmicb.2024.1416235] [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: 04/12/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Our previous studies have revealed that L. acidophilus possesses inhibitory effects on PCV2 proliferation in vivo, although the underlying mechanisms remain elusive. Probiotics like L. acidophilus are known to exert antiviral through their metabolites. Therefore, in this study, non-targeted metabolomics was used to detect the changes in metabolites of L. acidophilus after 24 h of proliferation. Subsequently, high-throughput molecular docking was utilized to analyze the docking scores of these metabolites with PCV2 Cap and Rep, aiming to identify compounds with potential anti-PCV2 effects. The results demonstrated that 128 compounds such as Dl-lactate were significantly increased. The results of high-throughput molecular docking indicated that compounds such as ergocristine, and telmisartan formed complexes with Cap and Rep, suggesting their potential anti-PCV2 properties. Furthermore, compounds like vitamin C, exhibit pharmacological effects consistent with L. acidophilus adding credence to the idea that L. acidophilus may exert pharmacological effects through its metabolites. These results will provide a foundation for the study of L. acidophilus.
Collapse
Affiliation(s)
- Zhigang Cao
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Xiaoya Ling
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Abdul Haseeb
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Panpan Sun
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Hua Zhang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Wei Yin
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Kuohai Fan
- Laboratory Animal Center, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Huizhen Yang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Zhenbiao Zhang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Jia Zhong
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Yaogui Sun
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Na Sun
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Hongquan Li
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| |
Collapse
|
20
|
Xiao C, Li X, Ding Z, Zhang H, Lv W, Yang C, He D, Zhu L. Enhancing Growth and Gut Health in Squabs: The Impact of Fermented Mixed Feed. Animals (Basel) 2024; 14:1411. [PMID: 38791629 PMCID: PMC11117316 DOI: 10.3390/ani14101411] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The purpose of this study was to evaluate the effect of fermented mixed feed (FMF) (soybean meal-rapeseed meal-corn bran (6:3:1, m/m/m)) on the growth performance, intestinal microbial communities, and metabolomes of squabs. One hundred and eighty 1-day-old squabs were randomly allocated to two groups, each containing six replicates of fifteen squabs cared for by 60 pairs of breeding pigeons secreting crop milk. Each pair of breeding pigeons cared for three squabs. The control group was fed a basal diet, while the experimental group was fed the basal diet containing 5% FMF. The results showed that daily weight gain, carcass weight, villus height, and the mRNA level of ZO-1 in the ileum were increased in the birds fed FMF compared to the control squabs (p < 0.05). Greater abundances of beneficial bacteria such as Lactobacillus, Bifidobacteria, and Bacillus as well as fewer harmful bacteria (i.e., Enterococcus, Veillonella, and Corynebacterium) in the ilea of squabs fed FMF. Six differential metabolites were identified in the FMF-treated squabs; one metabolite was increased (ω-salicoyisalicin) and five were decreased (3-benzoyloxy-6-oxo-12-ursen-28-oic acid, estradiol-17-phenylpropionate, aminotriazole, phosphatidyl ethanolamine (22:6/0:0), and 1-arachidonoylglycerophosphoinositol). Positive correlations were observed between the abundance of Lactobacillus and villus height. Overall, FMF treatment improved both growth and intestinal health in pigeons, suggesting potential benefits for pigeon production.
Collapse
Affiliation(s)
- Changfeng Xiao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Xin Li
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Zhizhao Ding
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Hongcai Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Wenwei Lv
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Changsuo Yang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Daqian He
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| | - Lihui Zhu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (C.X.); (X.L.); (Z.D.); (W.L.); (C.Y.); (D.H.)
| |
Collapse
|
21
|
Cao X, Wang B, Ren W, Wang J, Liu Q, Ren Y, Meng X. Mechanism of Ligilactobacillus salivarius GX118 in Regulating the Growth of Rainbow Trout (Oncorhynchus mykiss) and Resistance to Aeromonas salmonicida Infection. FISHES 2024; 9:157. [DOI: 10.3390/fishes9050157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Lactic acid bacteria Ligilactobacillus salivarius has been shown to be a substitute for antibiotics in the treatment of bacterial disease in high animals. However, its beneficial mechanism in fish farming is still unclear. This study evaluated the antagonistic effects of the Ligilactobacillus salivarius GX118 strain on Aeromonas salmonicida and its regulation of rainbow trout growth in vivo and in vitro. The results found that GX118 produces an antibacterial substance that can directly destroy the cell wall of A. salmonicida. Whole-genome sequencing of GX118 revealed that Enterolysin A is a type III bacteriocin with antibacterial properties. An in vivo experiment showed that the supplementation of GX118 in diet competitively inhibited the colonization of A. salmonicida in the intestine. In addition, it was able to improve the growth performance of rainbow trout within a 21-day feeding experiment. The supplementation of GX118 increased the diversity of gut microbiota, in which the abundance of Bacteroidota, Blautia, and Rhodobacteraceae increased. In addition, the use of GX118 activated the expression of IFN-γ and NF-κB genes and reduced the expression level of IL-6 and IL-8, thus exhibiting a certain effect on activating the immunity of rainbow trout. This study provides a scientific basis for the development of antibacterial probiotics in the healthy farming of rainbow trout.
Collapse
Affiliation(s)
- Xiyu Cao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Bowen Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Wenhao Ren
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Jiang Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Qi Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Yichao Ren
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Xianliang Meng
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266072, China
| |
Collapse
|
22
|
Pugazhendhi AS, Seal A, Hughes M, Kumar U, Kolanthai E, Wei F, Schwartzman JD, Coathup MJ. Extracellular Proteins Isolated from L. acidophilus as an Osteomicrobiological Therapeutic Agent to Reduce Pathogenic Biofilm Formation, Regulate Chronic Inflammation, and Augment Bone Formation In Vitro. Adv Healthc Mater 2024; 13:e2302835. [PMID: 38117082 DOI: 10.1002/adhm.202302835] [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: 08/25/2023] [Revised: 10/31/2023] [Indexed: 12/21/2023]
Abstract
Periprosthetic joint infection (PJI) is a challenging complication that can occur following joint replacement surgery. Efficacious strategies to prevent and treat PJI and its recurrence remain elusive. Commensal bacteria within the gut convey beneficial effects through a defense strategy named "colonization resistance" thereby preventing pathogenic infection along the intestinal surface. This blueprint may be applicable to PJI. The aim is to investigate Lactobacillus acidophilus spp. and their isolated extracellular-derived proteins (LaEPs) on PJI-relevant Staphylococcus aureus, methicillin-resistant S. aureus, and Escherichia coli planktonic growth and biofilm formation in vitro. The effect of LaEPs on cultured macrophages and osteogenic, and adipogenic human bone marrow-derived mesenchymal stem cell differentiation is analyzed. Data show electrostatically-induced probiotic-pathogen species co-aggregation and pathogenic growth inhibition together with LaEP-induced biofilm prevention. LaEPs prime macrophages for enhanced microbial phagocytosis via cathepsin K, reduce lipopolysaccharide-induced DNA damage and receptor activator nuclear factor-kappa B ligand expression, and promote a reparative M2 macrophage morphology under chronic inflammatory conditions. LaEPs also significantly augment bone deposition while abating adipogenesis thus holding promise as a potential multimodal therapeutic strategy. Proteomic analyses highlight high abundance of lysyl endopeptidase, and urocanate reductase. Further, in vivo analyses are warranted to elucidate their role in the prevention and treatment of PJIs.
Collapse
Affiliation(s)
| | - Anouska Seal
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | | | - Udit Kumar
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), University of Central Florida, Orlando, FL, 32826, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), University of Central Florida, Orlando, FL, 32826, USA
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | | | - Melanie J Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
23
|
Zhang F, Ni L, Zhang Z, Luo X, Wang X, Zhou W, Chen J, Liu J, Qu Y, Liu K, Guo L. Recombinant L. lactis vaccine LL-plSAM-WAE targeting four virulence factors provides mucosal immunity against H. pylori infection. Microb Cell Fact 2024; 23:61. [PMID: 38402145 PMCID: PMC10893618 DOI: 10.1186/s12934-024-02321-4] [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: 09/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) causes chronic gastric disease. An efficient oral vaccine would be mucosa-targeted and offer defense against colonization of invasive infection in the digestive system. Proteolytic enzymes and acidic environment in the gastrointestinal tract (GT) can, however, reduce the effectiveness of oral vaccinations. For the creation of an edible vaccine, L. lactis has been proposed as a means of delivering vaccine antigens. RESULTS We developed a plSAM (pNZ8148-SAM) that expresses a multiepitope vaccine antigen SAM-WAE containing Urease, HpaA, HSP60, and NAP extracellularly (named LL-plSAM-WAE) to increase the efficacy of oral vaccinations. We then investigated the immunogenicity of LL-plSAM-WAE in Balb/c mice. Mice that received LL-plSAM-WAE or SAM-WAE with adjuvant showed increased levels of antibodies against H. pylori, including IgG and sIgA, and resulted in significant reductions in H. pylori colonization. Furthermore, we show that SAM-WAE and LL-plSAM-WAE improved the capacity to target the vaccine to M cells. CONCLUSIONS These findings suggest that recombinant L. lactis could be a promising oral mucosa vaccination for preventing H. pylori infection.
Collapse
Affiliation(s)
- Furui Zhang
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Linhan Ni
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Zhang
- Department of Geriatrics and Special Needs Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xuequan Wang
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, 317000, China
| | - Wenmiao Zhou
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiale Chen
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jing Liu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Yuliang Qu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
| | - Kunmei Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
| | - Le Guo
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China.
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
24
|
De Santa F, Strimpakos G, Marchetti N, Gargari G, Torcinaro A, Arioli S, Mora D, Petrella C, Farioli-Vecchioli S. Effect of a multi-strain probiotic mixture consumption on anxiety and depression symptoms induced in adult mice by postnatal maternal separation. MICROBIOME 2024; 12:29. [PMID: 38369490 PMCID: PMC10875865 DOI: 10.1186/s40168-024-01752-w] [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: 03/27/2023] [Accepted: 01/04/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Intestinal microbial composition not only affects the health of the gut but also influences centrally mediated systems involved in mood, through the "gut-brain" axis, a bidirectional communication between gut microbiota and the brain. In this context, the modulation of intestinal microbiota and its metabolites through the administration of probiotics seems to represent a very promising approach in the treatment of the central nervous system alterations. Early postnatal life is a critical period during which the brain undergoes profound and essential modulations in terms of maturation and plasticity. Maternal separation (MS), i.e., the disruption of the mother-pup interaction, represents a pivotal paradigm in the study of stress-related mood disorders, by inducing persistent changes in the immune system, inflammatory processes, and emotional behavior in adult mammals. RESULTS We conducted experiments to investigate whether sustained consumption of a multi-strain probiotic formulation by adult male mice could mitigate the effects of maternal separation. Our data demonstrated that the treatment with probiotics was able to totally reverse the anxiety- and depressive-like behavior; normalize the neuro-inflammatory state, by restoring the resting state of microglia; and finally induce a proneurogenic effect. Mice subjected to maternal separation showed changes in microbiota composition compared to the control group that resulted in permissive colonization by the administered multi-strain probiotic product. As a consequence, the probiotic treatment also significantly affected the production of SCFA and in particular the level of butyrate. CONCLUSION Gut microbiota and its metabolites mediate the therapeutic action of the probiotic mix on MS-induced brain dysfunctions. Our findings extend the knowledge on the use of probiotics as a therapeutic tool in the presence of alterations of the emotional sphere that significantly impact on gut microbiota composition. Video Abstract.
Collapse
Affiliation(s)
- Francesca De Santa
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Georgios Strimpakos
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Nicole Marchetti
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
- Sciences of Nutrition, Aging, Metabolism and Gender Pathologies, Catholic University of Roma, Rome, 00100, Italy
| | - Giorgio Gargari
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Alessio Torcinaro
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy
| | - Stefania Arioli
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Diego Mora
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Policlinico Umberto I, Rome, Italy
| | - Stefano Farioli-Vecchioli
- Institute of Biochemistry and Cell Biology, IBBC, CNR, Via E. Ramarini, 32, Monterotondo, Rome, 00015, Italy.
| |
Collapse
|
25
|
Wang J, Liu Y, Guo Y, Liu C, Yang Y, Fan X, Yang H, Liu Y, Ma T. Function and inhibition of P38 MAP kinase signaling: Targeting multiple inflammation diseases. Biochem Pharmacol 2024; 220:115973. [PMID: 38103797 DOI: 10.1016/j.bcp.2023.115973] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Inflammation is a natural host defense mechanism that protects the body from pathogenic microorganisms. A growing body of research suggests that inflammation is a key factor in triggering other diseases (lung injury, rheumatoid arthritis, etc.). However, there is no consensus on the complex mechanism of inflammatory response, which may include enzyme activation, mediator release, and tissue repair. In recent years, p38 MAPK, a member of the MAPKs family, has attracted much attention as a central target for the treatment of inflammatory diseases. However, many p38 MAPK inhibitors attempting to obtain marketing approval have failed at the clinical trial stage due to selectivity and/or toxicity issues. In this paper, we discuss the mechanism of p38 MAPK in regulating inflammatory response and its key role in major inflammatory diseases and summarize the synthetic or natural products targeting p38 MAPK to improve the inflammatory response in the last five years, which will provide ideas for the development of novel clinical anti-inflammatory drugs based on p38 MAPK inhibitors.
Collapse
Affiliation(s)
- Jiahui Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongjian Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yushi Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cen Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoxiao Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Tao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
26
|
Yarlagadda T, Zhu Y, Snape N, Carey A, Bryan E, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. Lactobacillus rhamnosus dampens cytokine and chemokine secretion from primary human nasal epithelial cells infected with rhinovirus. J Appl Microbiol 2024; 135:lxae018. [PMID: 38268489 DOI: 10.1093/jambio/lxae018] [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: 12/03/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
AIMS To investigate the effect of Lactobacillus rhamnosus on viral replication and cellular response to human rhinovirus (HRV) infection, including the secretion of antiviral and inflammatory mediators from well-differentiated nasal epithelial cells (WD-NECs). METHODS AND RESULTS The WD-NECs from healthy adult donors (N = 6) were cultured in vitro, exposed to different strains of L. rhamnosus (D3189, D3160, or LB21), and infected with HRV (RV-A16) after 24 h. Survival and adherence capacity of L. rhamnosus in a NEC environment were confirmed using CFSE-labelled isolates, immunofluorescent staining, and confocal microscopy. Shed virus and viral replication were quantified using TCID50 assays and RT-qPCR, respectively. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity. Pro-inflammatory mediators were measured by multiplex immunoassay, and interferon (IFN)-λ1/3 was measured using a standard ELISA kit. Lactobacillus rhamnosus was able to adhere to and colonize WD-NECs prior to the RV-A16 infection. Lactobacillus rhamnosus did not affect shed RV-A16, viral replication, RV-A16-induced IFN-λ1/3 production, or LDH release. Pre-exposure to L. rhamnosus, particularly D3189, reduced the secretion of RV-A16-induced pro-inflammatory mediators by WD-NECs. CONCLUSIONS These findings demonstrate that L. rhamnosus differentially modulates RV-A16-induced innate inflammatory immune responses in primary NECs from healthy adults.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Yanshan Zhu
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Natale Snape
- University of Queensland Frazer Institute, Woolloongabba 4102, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| |
Collapse
|
27
|
Zha X, Su S, Wu D, Zhang P, Wei Y, Fan S, Huang Q, Peng X. The impact of gut microbiota changes on the intestinal mucus barrier in burned mice: a study using 16S rRNA and metagenomic sequencing. BURNS & TRAUMA 2023; 11:tkad056. [PMID: 38130728 PMCID: PMC10734567 DOI: 10.1093/burnst/tkad056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Background The gut microbiota is a complex ecosystem that plays a critical role in human health and disease. However, the relationship between gut microbiota and intestinal damage caused by burns is not well understood. The intestinal mucus layer is crucial for maintaining intestinal homeostasis and providing a physiological barrier against bacterial invasion. This study aims to investigate the impact of gut microbiota on the synthesis and degradation of intestinal mucus after burns and explore potential therapeutic targets for burn injury. Methods A modified histopathological grading system was employed to investigate the effects of burn injury on colon tissue and the intestinal mucus barrier in mice. Subsequently, 16S ribosomal RNA sequencing was used to analyze alterations in the gut microbiota at days 1-10 post-burn. Based on this, metagenomic sequencing was conducted on samples collected at days 1, 5 and 10 to investigate changes in mucus-related microbiota and explore potential underlying mechanisms. Results Our findings showed that the mucus barrier was disrupted and that bacterial translocation occurred on day 3 following burn injury in mice. Moreover, the gut microbiota in mice was significantly disrupted from days 1 to 3 following burn injury, but gradually recovered to normal as the disease progressed. Specifically, there was a marked increase in the abundance of symbiotic and pathogenic bacteria associated with mucin degradation on day 1 after burns, but the abundance returned to normal on day 5. Conversely, the abundance of probiotic bacteria associated with mucin synthesis changed in the opposite direction. Further analysis revealed that after a burn injury, bacteria capable of degrading mucus may utilize glycoside hydrolases, flagella and internalins to break down the mucus layer, while bacteria that synthesize mucus may help restore the mucus layer by promoting the production of short-chain fatty acids. Conclusions Burn injury leads to disruption of colonic mucus barrier and dysbiosis of gut microbiota. Some commensal and pathogenic bacteria may participate in mucin degradation via glycoside hydrolases, flagella, internalins, etc. Probiotics may provide short-chain fatty acids (particularly butyrate) as an energy source for stressed intestinal epithelial cells, promote mucin synthesis and accelerate repair of mucus layer.
Collapse
Affiliation(s)
- Xule Zha
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Panyang Zhang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
28
|
Faghfouri AH, Afrakoti LGMP, Kavyani Z, Nogourani ZS, Musazadeh V, Jafarlou M, Dehghan P. The role of probiotic supplementation in inflammatory biomarkers in adults: an umbrella meta-analysis of randomized controlled trials. Inflammopharmacology 2023; 31:2253-2268. [PMID: 37698776 DOI: 10.1007/s10787-023-01332-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE Despite the increasing evidence for probiotics' anti-inflammatory effects, the results of meta-analyses remain inconsistent. The present umbrella meta-analysis aimed to investigate the effects of probiotic supplementation on inflammatory biomarkers. METHODS We performed a wide-ranging systematic search in several databases, including PubMed, Web of Science, Scopus, EMBASE, and Google Scholar up to April 2023. The overall effect sizes were calculated using effect size (ES) values and their corresponding confidence intervals (CI). RESULTS Out of a total of 580 related articles, 39 studies were qualified for inclusion in the analysis. The results of the analysis revealed a significant reduction of C-reactive protein (CRP) (ES = -1.02; 95% CI: -1.23, -0.80, p < 0.001; I2: 94.1%, p < 0.001), TNF-α (ES = -0.35; 95% CI: -0.50, -0.20, p < 0.001; I2: 75.6%, p < 0.001), and interleukin-6 (IL-6) levels (ES = -0.36; 95% CI: -0.59, -0.13, p = 0.002; I2: 85.6%, p < 0.001), following probiotic supplementation. CONCLUSION Probiotic supplementation significantly reduced serum concentrations of TNF-a, CRP, and IL-6. Thus, probiotic supplementation can be considered adjuvant therapy to alleviate inflammation in various inflammatory conditions.
Collapse
Affiliation(s)
- Amir Hossein Faghfouri
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Zeynab Kavyani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vali Musazadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
- School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Jafarlou
- Faculty of Medicine and Health Science, UPM, 43400, Serdang, Malaysia
| | - Parvin Dehghan
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Li C, Peng K, Xiao S, Long Y, Yu Q. The role of Lactobacillus in inflammatory bowel disease: from actualities to prospects. Cell Death Discov 2023; 9:361. [PMID: 37773196 PMCID: PMC10541886 DOI: 10.1038/s41420-023-01666-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Inflammatory Bowel Disease (IBD), a chronic nonspecific intestinal inflammatory disease, is comprised of Ulcerative Colitis (UC) and Crohn's Disease (CD). IBD is closely related to a systemic inflammatory reaction and affects the progression of many intestinal and extraintestinal diseases. As one of the representative bacteria for probiotic-assisted therapy in IBD, multiple strains of Lactobacillus have been proven to alleviate intestinal damage and strengthen the intestinal immunological barrier, epithelial cell barrier, and mucus barrier. Lactobacillus also spares no effort in the alleviation of IBD-related diseases such as Colitis-associated Colorectal cancer (CAC), Alzheimer's Disease (AD), Depression, Anxiety, Autoimmune Hepatitis (AIH), and so on via gut-brain axis and gut-liver axis. This article aims to discuss the role of Lactobacillus in IBD and IBD-related diseases, including its underlying mechanisms and related curative strategies from the present to the future.
Collapse
Affiliation(s)
- Congxin Li
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kaixin Peng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Siqi Xiao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Yuanyuan Long
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Qin Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
30
|
Rodenes-Gavidia A, Lamelas A, Bloor S, Hobson A, Treadway S, Haworth J, Vijayakumar V, Naghibi M, Day R, Chenoll E. An insight into the functional alterations in the gut microbiome of healthy adults in response to a multi-strain probiotic intake: a single arm open label trial. Front Cell Infect Microbiol 2023; 13:1240267. [PMID: 37841999 PMCID: PMC10570534 DOI: 10.3389/fcimb.2023.1240267] [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: 06/14/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Background Probiotic supplements, by definition, provide a benefit to the host, but few studies have investigated the effect of probiotic supplements in healthy adult populations. Purpose The present, single arm, open label clinical trial, evaluated compositional and functional changes in the fecal microbiome of healthy adults after supplementation with a 14-strain probiotic. Methods We analysed the effect of a 14-strain probiotic blend (Bacillus subtilis NCIMB 30223, Bifidobacterium bifidum NCIMB 30179, B. breve NCIMB 30180, B. infantis NCIMB 30181, B. longum NCIMB 30182, Lactobacillus helveticus NCIMB 30184, L. delbrueckii subsp. bulgaricus NCIMB 30186, Lacticaseibacillus paracasei NCIMB 30185, Lactiplantibacillus plantarum NCIMB 30187, Lacticaseibacillus rhamnosus NCIMB 30188, L. helveticus NCIMB 30224, Lactobacillus salivarius NCIMB 30225, Lactococcus lactis subsp. lactis NCIMB 30222, and Streptococcus thermophilus NCIMB 30189), on the faecal microbiota of healthy young adults (n=41) in a single arm study. The adults consumed 4 capsules daily of the 14 strain blend(8 billion colony forming units/day) for 8 weeks. Compositional and functional changes in faecal microbiota before and after supplementation were assessed using shotgun metagenomic sequencing. Fasting breath analysis, faecal biochemistry and bowel habits were also assessed. Results In healthy adult participants, no significant changes to the overall alpha- or beta-diversity was observed after 8 weeks of multi-strain probiotic supplementation. However, in a simplified model that considered only time and individual differences, significant decreases (p < 0.05) in family Odoribacteraceae and Bacteroidaceae abundance and a significant increase (p < 0.05) in genus Megamonas abundance were observed. At a functional level, there were significant changes in functional gene abundance related to several functional pathways, including phenylalanine metabolism, O-antigen nucleotide sugar biosynthesis, bacterial chemotaxis, and flagellar assembly. No significant changes in stool form or frequency, fecal biochemistry, or methane and hydrogen breath tests were observed. Conclusion In healthy young adults, overall alpha- and beta-diversity did not change in response to probiotic intake even though modest compositional changes at the family and genus level were observed. However, at functional level, results identified changes in gene abundance for several functional pathways.
Collapse
Affiliation(s)
- Andrea Rodenes-Gavidia
- ADM BIOPOLIS, University of Valencia Science Park (Parc Científic de la Universitat de València), Valencia, Spain
| | - Araceli Lamelas
- ADM BIOPOLIS, University of Valencia Science Park (Parc Científic de la Universitat de València), Valencia, Spain
| | - Sarah Bloor
- Functional Gut Clinic, Manchester, United Kingdom
- Anglia Ruskin University, Essex, Norwich, United Kingdom
| | - Anthony Hobson
- Functional Gut Clinic, Manchester, United Kingdom
- Anglia Ruskin University, Essex, Norwich, United Kingdom
| | - Sam Treadway
- Functional Gut Clinic, Manchester, United Kingdom
| | | | | | - Malwina Naghibi
- Medical Department, ADM Health & Wellness, Somerset, United Kingdom
| | - Richard Day
- Medical Department, ADM Health & Wellness, Somerset, United Kingdom
| | - Empar Chenoll
- ADM BIOPOLIS, University of Valencia Science Park (Parc Científic de la Universitat de València), Valencia, Spain
| |
Collapse
|
31
|
Li Y, Tong T, Li P, Peng Y, Zhang M, Liu J, She Y, Li Z, Li Y. Screening of Potential Probiotic Lactobacillaceae and Their Improvement of Type 2 Diabetes Mellitus by Promoting PI3K/AKT Signaling Pathway in db/db Mice. Pol J Microbiol 2023; 72:285-297. [PMID: 37725896 PMCID: PMC10508973 DOI: 10.33073/pjm-2023-028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/09/2023] [Indexed: 09/21/2023] Open
Abstract
The study aimed to isolate Lactobacillaceae strains with in vitro hypoglycemic activity and probiotic properties and to determine their antidiabetic abilities in vivo. Lactiplantibacillus plantarum 22, L. plantarum 25, Limosilactobacillus fermentum 11, and L. fermentum 305 with high in vitro hypoglycemic activity were screened from 23 strains of Lactobacillaceae isolated from human feces and identified by 16S rDNA sequencing. The fasting blood glucose (FBG) of the mice was recorded weekly. After 12 weeks, liver, kidney, and pancreas tissues were stained with hematoxylin and eosin (H&E) to observe histomorphology; the inflammatory factors were assayed by Quantitative Real-time PCR; PI3K and AKT were measured by Western blot; the short-chain fatty acids (SCFAs) were determined by LC-MS/MS. Inhibitory activities of L. plantarum 22, L. plantarum 25, L. fermentum 11, and L. fermentum 305 against α-amylase were 62.29 ± 0.44%, 51.81 ± 3.65%, 58.40 ± 1.68%, and 57.48 ± 5.04%, respectively. Their inhibitory activities to α-glucosidase were 14.89 ± 0.38%, 15.32 ± 0.89%, 52.63 ± 3.07%, and 51.79 ± 1.13%, respectively. Their survival rate after simulated gastrointestinal test were 12.42 ± 2.84%, 9.10 ± 1.12%, 5.86 ± 0.52%, and 8.82 ± 2.50% and their adhesion rates to Caco-2 cell were 6.09 ± 0.39%, 6.37 ± 0.28%, 6.94 ± 0.27%, and 6.91 ± 0.11%, respectively. The orthogonal tests of bacterial powders of the four strains showed that the maximum inhibitory activities to α-amylase and α-glucosidase were 93.18 ± 1.19% and 75.33 ± 2.89%, respectively. The results showed that the mixture of Lactobacillaceae could lower FBG, reduce inflammation, and liver, kidney, and pancreas damage, promote PI3K/AKT signaling pathway, and increase the content of SCFAs. The combination of L. plantarum 22, L. plantarum 25, L. fermentum 11, and L. fermentum 305 can potentially improve type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Yueyang Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Tong Tong
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Peifan Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yian Peng
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- School of Public Health, Anhui University of Science and Technology, Hefei, China
| | - Jia Liu
- Internal Trade Food Science and Technology (Beijing) Co., Ltd., Beijing, China
| | - Yongxin She
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Zuming Li
- College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yongli Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Zhao J, Liao Y, Wei C, Ma Y, Wang F, Chen Y, Zhao B, Ji H, Wang D, Tang D. Potential Ability of Probiotics in the Prevention and Treatment of Colorectal Cancer. Clin Med Insights Oncol 2023; 17:11795549231188225. [PMID: 37601319 PMCID: PMC10437046 DOI: 10.1177/11795549231188225] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 08/22/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world, and its incidence rate and mortality are on the rise in many countries. In recent years, with the improvement of economic conditions, people's living habits have changed, including lack of physical activity, poor diet patterns and circadian rhythm disorder. These risk factors can change the colon environment and the composition of intestinal microbiota. This state is called intestinal imbalance, which increases the risk of cancer. Probiotics, a class of microorganisms that help maintain gut microbial homeostasis and alleviate dysbiosis, may help prevent inflammation and colorectal cancer. These probiotics inhibit or ameliorate the effects of dysbiosis through the production of short-chain fatty acids (SCFAs), modulation of immunity, maintenance of the intestinal epithelial barrier, pro-apoptotic mechanisms, and other mechanisms. This review aims to explain the interaction between probiotics, the gut microenvironment and the gut microbiota, and summarize reports on the possibility of probiotics in the prevention and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Jiahao Zhao
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yiqun Liao
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Chen Wei
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Yichao Ma
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Fei Wang
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Yuji Chen
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Bin Zhao
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Hao Ji
- Clinical Medical College, Dalian Medical University, Dalian, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou, China
| |
Collapse
|
33
|
Musazadeh V, Nazari A, Faghfouri AH, Emami M, Kavyani Z, Zokaei M, Jamilian P, Zarezadeh M, Saedisomeolia A. The effectiveness of treatment with probiotics in Helicobacter pylori eradication: results from an umbrella meta-analysis on meta-analyses of randomized controlled trials. Food Funct 2023; 14:7654-7662. [PMID: 37540067 DOI: 10.1039/d3fo00300k] [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: 08/05/2023]
Abstract
Background and aims: The purpose of this umbrella meta-analysis was to quantitatively summarize meta-analyses of randomized controlled trial (RCT) studies regarding the effects of probiotic supplementation on Helicobacter pylori (H. pylori) eradication. Methods: A thorough search of the electronic databases including PubMed, Web of Science, Embase, Scopus, and Google Scholar was carried out from the inception up to May 2022. For the evaluation of overall effect sizes, the pooled relative risk (RR) or odds ratio (OR) and their corresponding 95% confidence intervals (CI) were calculated. The random-effects model was used for the meta-analysis. Results: Overall, 18 eligible studies (47 278 participants in total) were included in the study. The findings revealed that probiotics have a beneficial impact on H. pylori eradication (pooled ESRR: 1.13; 95% CI: 1.11, 1.14, p < 0.01, and ESOR = 1.86, 95% CI: 1.70, 2.03, p < 0.01). Greater effects on H. pylori eradication were observed when higher doses (>10 × 1010 CFU) and mixed strains were supplemented. Conclusion: The present umbrella meta-analysis suggests that supplementation with probiotics may be considered as an efficient approach to ameliorate H. pylori complications, particularly probiotics with higher CFUs and mixed strains.
Collapse
Affiliation(s)
- Vali Musazadeh
- Student Research Committee, Tabriz University of Medical, Sciences, Tabriz, Iran
- School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Faghfouri
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Emami
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Kavyani
- Student Research Committee, Tabriz University of Medical, Sciences, Tabriz, Iran
- School of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parmida Jamilian
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| | - Meysam Zarezadeh
- Student Research Committee, Tabriz University of Medical, Sciences, Tabriz, Iran
- Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Attar-Neishaburi St., Golgasht Alley, Azadi Blvd., Tabriz, Iran.
| | - Ahmad Saedisomeolia
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, 21, 111 Lakeshore, Ste-Anne-de-Bellevue, Quebec, H9X 3 V9, Canada.
| |
Collapse
|
34
|
Li J, Feng S, Pi Y, Jiang X, Li X, Zhou Z, Liu X, Wei H, Tao S. Limosilactobacillus johnsoni and Limosilactobacillus mucosae and Their Extracellular Vesicles Alleviate Gut Inflammatory Injury by Mediating Macrophage Polarization in a Lipopolysaccharide-Challenged Piglet Model. J Nutr 2023; 153:2497-2511. [PMID: 37343627 DOI: 10.1016/j.tjnut.2023.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Limosilactobacillus johnsoni (L. j) and Limosilactobacillus mucosae (L. m) can alleviate the inflammatory response. OBJECTIVES This study aimed to elucidate the underlying mechanisms by which L. j- and L. m-derived extracellular vesicles (EVs) mitigate lipopolysaccharide (LPS)-induced intestinal injury. METHODS Piglets were assigned to 4 groups: oral phosphate-buffered saline inoculation for 2 wk prior to intraperitoneal injection of physiological saline or LPS, and oral L. j/L. m inoculation for 2 wk prior to intraperitoneal injection of LPS. The intestinal integrity, macrophage markers, cytokine levels, and microbiota were determined. The cytokine levels and macrophage phenotype were detected after L. j/L. m and their EVs were coincubated with macrophages. The levels of cytokines, tight junction proteins, and apoptosis were measured after intestinal epithelial cells were cocultured with macrophages. RESULTS LPS challenge decreased jejunal villus length; expression levels of zonula occludens-1 (ZO-1), occludin, arginase-1 (Arg1), and interleukin (IL)-10; and number of CD163+ cells and increased the expression levels of inducible nitric oxide synthase (iNOS), IL-1β, IL-6, and tumor necrosis factor (TNF)-α compared with that in the control. L. j and L. m pretreatment rescued the aforementioned indicators compared with LPS challenge. Pretreatment of L. j and L. m and their EVs reversed the levels of IL-1β, IL-6, TNF-α, and IL-10 and the gene expression of iNOS and Arg1 in the LPS group in macrophages. Pretreatment with L. j and L. m-derived EVs increased ZO-1 and occludin mRNA expression and reduced IL-1β, caspase-3, and bax gene expression in intestinal epithelial cells of the coculture system. Enzyme-treated EVs were less effective than native EVs. CONCLUSIONS This study suggests that EVs secreted by L. j and L. m control inflammation by modulating macrophage polarization, thereby improving intestinal barrier function.
Collapse
Affiliation(s)
- Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shengkai Feng
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yu Pi
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xianren Jiang
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zutao Zhou
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
35
|
Sun Q, Yin S, He Y, Cao Y, Jiang C. Biomaterials and Encapsulation Techniques for Probiotics: Current Status and Future Prospects in Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2185. [PMID: 37570503 PMCID: PMC10421492 DOI: 10.3390/nano13152185] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023]
Abstract
Probiotics have garnered significant attention in recent years due to their potential advantages in diverse biomedical applications, such as acting as antimicrobial agents, aiding in tissue repair, and treating diseases. These live bacteria must exist in appropriate quantities and precise locations to exert beneficial effects. However, their viability and activity can be significantly impacted by the surrounding tissue, posing a challenge to maintain their stability in the target location for an extended duration. To counter this, researchers have formulated various strategies that enhance the activity and stability of probiotics by encapsulating them within biomaterials. This approach enables site-specific release, overcoming technical impediments encountered during the processing and application of probiotics. A range of materials can be utilized for encapsulating probiotics, and several methods can be employed for this encapsulation process. This article reviews the recent advancements in probiotics encapsulated within biomaterials, examining the materials, methods, and effects of encapsulation. It also provides an overview of the hurdles faced by currently available biomaterial-based probiotic capsules and suggests potential future research directions in this field. Despite the progress achieved to date, numerous challenges persist, such as the necessity for developing efficient, reproducible encapsulation methods that maintain the viability and activity of probiotics. Furthermore, there is a need to design more robust and targeted delivery vehicles.
Collapse
Affiliation(s)
- Qiqi Sun
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan 250117, China; (Q.S.); (S.Y.)
| | - Sheng Yin
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan 250117, China; (Q.S.); (S.Y.)
- Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yingxu He
- School of Computing, National University of Singapore, Singapore 119077, Singapore;
| | - Yi Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan 250117, China; (Q.S.); (S.Y.)
- Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Chunping Jiang
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan 250117, China; (Q.S.); (S.Y.)
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210000, China
| |
Collapse
|
36
|
Ali MS, Lee EB, Hsu WH, Suk K, Sayem SAJ, Ullah HMA, Lee SJ, Park SC. Probiotics and Postbiotics as an Alternative to Antibiotics: An Emphasis on Pigs. Pathogens 2023; 12:874. [PMID: 37513721 PMCID: PMC10383198 DOI: 10.3390/pathogens12070874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Probiotics are being used as feed/food supplements as an alternative to antibiotics. It has been demonstrated that probiotics provide several health benefits, including preventing diarrhea, irritable bowel syndrome, and immunomodulation. Alongside probiotic bacteria-fermented foods, the different structural components, such as lipoteichoic acids, teichoic acids, peptidoglycans, and surface-layer proteins, offer several advantages. Probiotics can produce different antimicrobial components, enzymes, peptides, vitamins, and exopolysaccharides. Besides live probiotics, there has been growing interest in consuming inactivated probiotics in farm animals, including pigs. Several reports have shown that live and killed probiotics can boost immunity, modulate intestinal microbiota, improve feed efficiency and growth performance, and decrease the incidence of diarrhea, positioning them as an interesting strategy as a potential feed supplement for pigs. Therefore, effective selection and approach to the use of probiotics might provide essential features of using probiotics as an important functional feed for pigs. This review aimed to systematically investigate the potential effects of lactic acid bacteria in their live and inactivated forms on pigs.
Collapse
Affiliation(s)
- Md Sekendar Ali
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong 4318, Bangladesh
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Walter H Hsu
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50014, USA
| | - Kyoungho Suk
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Syed Al Jawad Sayem
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - H M Arif Ullah
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Seung-Jin Lee
- Development and Reproductive Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
37
|
Najafi S, Sotoodehnejadnematalahi F, Amiri MM, Pourshafie MR, Rohani M. Prophylactic vs. Therapeutic Effect of Probiotics on the Inflammation Mediated by the NF-κB Pathway in Inflammatory Bowel Conditions. Biomedicines 2023; 11:1675. [PMID: 37371769 DOI: 10.3390/biomedicines11061675] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/23/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Probiotic supplements consumed adequately at the proper time can affect health by modulating inflammatory pathways in gastrointestinal epithelial cells and modifying the resultant inflammatory response. The current study applied in vitro models to investigate the effectiveness of probiotics in modulating inflammatory pathways and altering inflammatory gene expression in gastrointestinal epithelial cells, with the ultimate goal of promoting probiotic consumption as a therapeutic and preventive measure for chronic inflammatory bowel conditions. HT-29 cells were treated with Gram-negative bacteria to evaluate the changes in pathways related to inflammation activities before and after treatment with a Lactobacillus spp. cocktail (L. plantarum, L. rhamnosus, L. brevis, and L. ruteri) and a Bifidobacterium spp. cocktail (B. bifidum, B. langum, and B. breve) using the real-time PCR method and ELISA for IL-1β and IL-6 as pro-inflammatory cytokines. The results showed that the expression of NF-κB signaling pathway genes and IL-1β and IL-6 cytokines increased after exposure to Gram-negative components. In contrast, all probiotic combinations significantly decreased the expression of genes and the secretion of cytokines. However, this decrease was significantly smaller in cells that underwent probiotic treatment after inflammation induction. In addition, cocktails containing combined Lactobacillus and Bifidobacterium demonstrated robust anti-inflammatory activity relative to solo cocktails. Our observations confirm that probiotic consumption could positively impact inflammatory conditions and alleviate inflammatory symptoms; they can be particularly effective as a preventive measure. Our study provides preliminary evidence to support the lifetime consumption of probiotics.
Collapse
Affiliation(s)
- Saeideh Najafi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | | | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran 14155-6619, Iran
| | | | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran 13169-43551, Iran
| |
Collapse
|
38
|
H S R, Halami PM. The Combined Effect of Potential Probiotic Bacillus licheniformis MCC 2514 and Bifidobacterium breve NCIM 5671 Towards Anti-inflammatory Activity on HT-29 Cell Lines. Probiotics Antimicrob Proteins 2023; 15:351-362. [PMID: 34581975 DOI: 10.1007/s12602-021-09851-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 12/23/2022]
Abstract
Probiotics are considered a natural source for treating many intestinal disorders, which deliver health benefits in different ways. The study aims to evaluate the immunomodulatory gene expression on HT-29 cell line using Bacillus licheniformis MCC 2514 and Bifidobacterium breve NCIM 5671 as a single culture and in combination. Upon inflammation induced by LPS, the combination of bacteria downregulated the pro-inflammatory cytokines IL-1α (13.4), IL-12 (14.6), IL-8 (2.6), and IL-6 (1.9), and in contrast, TNF-α (21.2) folds has upregulated. However, anti-inflammatory genes such as IL-4 (0.6), IL-10 (2.9), TGF-2 (92.2), and TGF-3 (85.8) folds were upregulated. The combination of bacteria against oxidative stress downregulated the pro-inflammatory cytokines such as IL-1α & β, IL-6, IL-8, IL-12, and IL-18, and upregulated the anti-inflammatory cytokines IL-10, IL-4, TGF-2, and TGF-3. On the introduction of Kocuria rhizophila, the pro-inflammatory cytokines were upregulated. On supplementation of B. licheniformis and B. breve, the upregulated pro-inflammatory cytokines were decreased, and anti-inflammatory cytokines such as IL-4 (6.2), IL-10 (23.5), TGF-2 (166), and TGF-3(28.4) folds were increased. However, gene expression of toll-like receptor-2 was found high (26 folds) upon introducing probiotic bacteria. ELISA results of Interferon-γ found that the expression was higher (7.19 ng/mL) on the introduction of both the bacteria in combination. The higher anti-inflammatory activity was observed when potential probiotic bacteria were used in combination compared to a single culture. Overall study indicates that the combination of aerobic B. licheniformis and anaerobic B. breve has an anti-inflammatory activity that can sustain an excellent gastrointestinal environment during pathogen invasion and inflammation.
Collapse
Affiliation(s)
- Rohith H S
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Prakash Motiram Halami
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, India.
| |
Collapse
|
39
|
Zheng T, Wu Y, Guo KX, Tan ZJ, Yang T. The process of hypertension induced by high-salt diet: Association with interactions between intestinal mucosal microbiota, and chronic low-grade inflammation, end-organ damage. Front Microbiol 2023; 14:1123843. [PMID: 36925479 PMCID: PMC10011071 DOI: 10.3389/fmicb.2023.1123843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/27/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation and immunity play a major role in the development of hypertension, and a potential correlation between host mucosal immunity and inflammatory response regulation. We explored the changes of intestinal mucosal microbiota in hypertensive rats induced by high-salt diet and the potential link between the intestinal mucosal microbiota and inflammation in rats. Therefore, we used PacBio (Pacific Bioscience) SMRT sequencing technology to determine the structure of intestinal mucosal microbiota, used enzyme-linked immunosorbent assay (ELISA) to determined the proinflammatory cytokines and hormones associated with hypertension in serum, and used histopathology methods to observe the kidney and vascular structure. We performed a potential association analysis between intestinal mucosal characteristic bacteria and significantly different blood cytokines in hypertensive rats induced by high-salt. The results showed that the kidney and vascular structures of hypertensive rats induced by high salt were damaged, the serum concentration of necrosis factor-α (TNF-α), angiotensin II (AngII), interleukin-6 (IL-6), and interleukin-8 (IL-8) were significantly increased (p < 0.05), and the coefficient of immune organ spleen was significantly changed (p < 0.05), but there was no significant change in serum lipids (p > 0.05). From the perspective of gut microbiota, high-salt diet leads to significant changes in intestinal mucosal microbiota. Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were the dominant differential bacteria in intestinal mucosal, with the AUC (area under curve) value of Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were 1 and 0.875 according to ROC (receiver operating characteristic) analysis. Correlation analysis showed that Bifidobacterium animalis subsp. was correlated with IL-6, IL-8, TNF-α, and Ang II. Based on our results, we can speculated that high salt diet mediated chronic low-grade inflammation through inhibited the growth of Bifidobacterium animalis subsp. in intestinal mucosa and caused end-organ damage, which leads to hypertension.
Collapse
Affiliation(s)
- Tao Zheng
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Wu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kang-xiao Guo
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Zhou-jin Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Tao Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
40
|
Jing N, Liu F, Wang R, Zhang Y, Yang J, Hou Y, Zhang H, Xie Y, Liu H, Ge S, Jin J. Both live and heat-killed Bifidobacterium animalis J-12 alleviated oral ulcers in LVG golden Syrian hamsters by gavage by directly intervening in the intestinal flora structure. Food Funct 2023; 14:2045-2058. [PMID: 36723265 DOI: 10.1039/d2fo03751c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Live and heat-killed Bifidobacterium has been proven to have anti-inflammatory and antioxidant effects. In this study, we evaluated the effects of live and heat-killed Bifidobacterium animalis J-12 (J-12) on the oral ulceration of LVG golden Syrian hamsters after buccal membrane injection with methyl viologen dichloride. Results showed that interleukin-1β, glutathione, and malondialdehyde in serum were downregulated by the gavage of live and heat-killed J-12 bacteria. The J-12 live and heat-killed bacteria can reduce the expression of matrix metalloproteinase-9 by reducing the expression of nuclear factor kappa-B, thus reducing the expression of anti-inflammatory factors lipoxin A4 and prostaglandin E2. Reducing the expression of caspase-3 and adenosine diphosphate ribose polymerase resulted in a reduction of ulcer tissue DNA damage. In addition, regulating the structure of the intestinal flora prevented the process of oral ulcer formation. This study shows that J-12 can reduce the risk of oral ulcer formation while also having a positive effect on inhibiting existing oral ulcer growth.
Collapse
Affiliation(s)
- Nanqing Jing
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Fudong Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, Inner Mongolia 010110, China
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, Inner Mongolia 010110, China
| | - Ran Wang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100190, China
| | - Yan Zhang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Jianjun Yang
- Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Yubing Hou
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Hongxing Zhang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Yuanhong Xie
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Hui Liu
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| | - Shaoyang Ge
- BEIJING HEYIYUAN BIOTECHNOLOGY Co, Ltd., Beijing 100088, China
| | - Junhua Jin
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing Laboratory of Food Quality and Safety, Beijing Key Laboratory of Detection and Control of Spoilage Organisms and Pesticide Residues in Agricultural Products, Beijing 102206, China.
| |
Collapse
|
41
|
Chen J, Yan F, Kuttappan VA, Wedekind K, Vázquez-Añón M, Hancock D. Effects of bis-chelated copper in growth performance and gut health in broiler chickens subject to coccidiosis vaccination or coccidia challenge. Front Physiol 2023; 13:991318. [PMID: 36817619 PMCID: PMC9936238 DOI: 10.3389/fphys.2022.991318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/28/2022] [Indexed: 02/05/2023] Open
Abstract
Copper (Cu) is widely used at high levels as growth promoter in poultry, the alternative source of Cu to replace the high level of inorganic Cu at poultry farm remains to be determined. Three floor pen experiments were conducted to evaluate the effects of Cu methionine hydroxy-analogue chelate (Cu-MHAC, MINTREX®Cu, Novus International, Inc.) on growth performance and gut health in broilers in comparison to CuSO4 and/or tribasic copper chloride (TBCC). There were 3 treatments in experiment#1 (0, 30 and 75 ppm Cu-MHAC) and experiment#2 (15 and 30 ppm Cu-MHAC, and 125 ppm CuSO4), and 4 treatments in experiment #3 (15 and 30 ppm Cu-MHAC, 125 ppm CuSO4 and 125 ppm TBCC) with nine replicates pens of 10-13 birds in each treatment. The levels of other minerals were equal among all treatments within each experiment. All birds were orally gavaged with a coccidiosis vaccine at 1x recommended dose on d0 in experiment#1 and #2 and 10x recommended dose on d15 in experiment #3. Data were analyzed by one-way ANOVA, means were separated by Fisher's protected LSD test. A p ≤ 0.05 was considered statistically different. In experiment #1, 30 and 75 ppm Cu-MHAC improved FCR during grower phase, increased jejunal villus height and reduced jejunal crypt depth, 30 ppm Cu-MHAC increased cecal Lactobacillus spp. abundance in 41 days broilers. In experiment #2, compared to CuSO4, 15ppm Cu-MHAC increased cumulative performance index in 28 days broilers, 15 and/or 30 ppm Cu-MHAC improved gut morphometry, and 30 ppm Cu-MHAC reduced the abundance of E. coli and Enterobacteriaceae in cecum in 43 days broilers. In experiment #3, 15 ppm and 30 ppm Cu-MHAC improved FCR vs. CuSO4 during starter phase, reduced the percentage of E. coli of total bacteria vs. TBCC, 30 ppm Cu-MHAC increased the percentages of Lactobacillus acidophilus, Lactobacillus spp. and Clostridium cluster XIVa of total bacteria vs. both CuSO4 and TBCC in the cecum of 27 days broilers. In summary, low doses of Cu-MHAC had comparable growth performance to high dose of TBCC and CuSO4 while improving gut microflora and gut morphometry in broilers subject to coccidiosis vaccination or coccidia challenge, indicating that low doses of bis-chelated Cu could be used as a complimentary strategy to improve animal gut health.
Collapse
|
42
|
The Role of the Gut Microbiome and Trimethylamine Oxide in Atherosclerosis and Age-Related Disease. Int J Mol Sci 2023; 24:ijms24032399. [PMID: 36768722 PMCID: PMC9917289 DOI: 10.3390/ijms24032399] [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: 01/06/2023] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
The gut microbiome plays a major role in human health, and gut microbial imbalance or dysbiosis is associated with disease development. Modulation in the gut microbiome can be used to treat or prevent different diseases. Gut dysbiosis increases with aging, and it has been associated with the impairment of gut barrier function leading to the leakage of harmful metabolites such as trimethylamine (TMA). TMA is a gut metabolite resulting from dietary amines that originate from animal-based foods. TMA enters the portal circulation and is oxidized by the hepatic enzyme into trimethylamine oxide (TMAO). Increased TMAO levels have been reported in elderly people. High TMAO levels are linked to peripheral artery disease (PAD), endothelial senescence, and vascular aging. Emerging evidence showed the beneficial role of probiotics and prebiotics in the management of several atherogenic risk factors through the remodeling of the gut microbiota, thus leading to a reduction in TMAO levels and atherosclerotic lesions. Despite the promising outcomes in different studies, the definite mechanisms of gut dysbiosis and microbiota-derived TMAO involved in atherosclerosis remain not fully understood. More studies are still required to focus on the molecular mechanisms and precise treatments targeting gut microbiota and leading to atheroprotective effects.
Collapse
|
43
|
Shen S, Yang W, Li L, Zhu Y, Yang Y, Ni H, Jiang Z, Zheng M. In vitro fermentation of seaweed polysaccharides and tea polyphenol blends by human intestinal flora and their effects on intestinal inflammation. Food Funct 2023; 14:1133-1147. [PMID: 36594623 DOI: 10.1039/d2fo03390a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A combination of polysaccharides and tea polyphenols can enhance immune activity synergistically, depending on the type and structure of polysaccharides, but the mechanism remains unknown. This study is aimed to investigate the regulating effects of different seaweed polysaccharide (ι-carrageenan, agarose) and tea polyphenol blends on intestinal flora and intestinal inflammation using an in vitro ascending-transverse-descending colon fermentation system and RAW264.7 cell model. The results showed that seaweed polysaccharides in the presence of tea polyphenol were almost completely degraded at transverse colon fermentation for 36 h. Agarose significantly enhanced the butyric acid production content by increasing the abundance of Lachnospiraceae, whereas agarose and tea polyphenol blends did not have a synergistic effect. On the contrary, ι-carrageenan and tea polyphenol blends synergistically increased the abundance of beneficial bacteria (e.g., Bacteroidetes and Bifidobacterium) and promoted the production of short-chain fatty acids (SCFAs), such as isobutyric acid. Such changes tended to alter the impacts of different seaweed polysaccharides and tea polyphenol blends on intestinal inflammation. Among them, ι-carrageenan and tea polyphenol blends were the most effective in inhibiting lipopolysaccharide-induced NO, ROS, IL-6, and TNF-α production in RAW264.7 cells, indicating the alleviated intestinal inflammation. The results suggest that the seaweed polysaccharide and tea polyphenol blends have prebiotic potential and can benefit intestinal health.
Collapse
Affiliation(s)
- Shiqi Shen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China.
| | - Wenqin Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China.
| | - Lijun Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Yanbing Zhu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Yuanfan Yang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Hui Ni
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China.,Xiamen Ocean Vocational College, Xiamen 361100, Fujian, China
| | - Zedong Jiang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Mingjing Zheng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China. .,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, Liaoning 116034, China.,Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| |
Collapse
|
44
|
Chopra H, Goyal R, Baig AA, Arora S, Dua K, Gautam RK. Synbiotics in Colon Cancer. SYNBIOTICS FOR THE MANAGEMENT OF CANCER 2023:115-133. [DOI: 10.1007/978-981-19-7550-9_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
45
|
Liu J, Tao Y, Haikun W, Lanfang Y, Jingyi L, Ping Y. Triclosan exposure induced disturbance of gut microbiota and exaggerated experimental colitis in mice. BMC Gastroenterol 2022; 22:469. [DOI: 10.1186/s12876-022-02527-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/05/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Background
Triclosan, an antimicrobial agent in personal care products, could be absorbed into the human body through the digestive tract. This animal experiment aimed to clarify the effects of triclosan exposure on the microbiome and intestinal immune functions in healthy and ulcerative colitis models.
Methods
Balb/c mice were maintained on an AIN-93G diet containing 80ppm triclosan dissolved in polyethylene as vehicle or vehicle alone for 1 week or 4 weeks. In the end, the mice were sacrificed, blood samples and colon tissues were collected for analysis of inflammation, and fecal samples were collected for 16 S rRNA sequencing of gut microbiota. To establish ulcerative colitis mice model, at the beginning of the 4th week, mice maintained on the diet with or without triclosan were treated with 2% Dextran sulfate sodium(DSS) in drinking water for 1 week. Then mice were sacrificed for analysis of colitis and gut microbiota.
Results
Triclosan exposure to common mice enhanced the levels of p-NF-κb and Toll-like receptor 4 (TLR4), and decreased the Occludin in the colon. Triclosan exposure to DSS-induced mice increased the level of inflammatory cytokines, reduced the levels of Occludin, and exacerbated the degree of damage to intestinal mucosa and crypt, infiltration of inflammatory cells and atypia of glandular cells. Low-grade intraepithelial neoplasia appeared. Both in common and DSS-induced mice, triclosan exposure changed the diversity and composition of gut microbiota. Fecal samples showed higher enrichment of sulfate-reducing bacteria and Bacteroides, and less butyrate-producing bacteria.
Conclusion
Triclosan exposure induced disturbance of gut microbiota and exaggerated experimental colitis in mice. And changes in the composition of gut microbiota were characterized by the increase of harmful bacteria, including sulfate-reducing bacteria and Bacteroides, and the reduction of protective probiotics, butyrate-producing bacteria.
Collapse
|
46
|
Salem GA, Mohamed AAR, Ghonimi WAM, Abdallah HM, Rhouma NR, Ali RI. The synbiotic mixture of Bacillus licheniformis and Saccharomyces cerevisiae extract aggravates dextran sulfate sodium induced colitis in rats. BMC Vet Res 2022; 18:405. [PMID: 36384756 PMCID: PMC9667625 DOI: 10.1186/s12917-022-03479-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Uncertain effects of probiotics and/or prebiotics have been reported in experimental and clinical colitis. This study aims to examine the effects of a synbiotic combination comprising Bacillus licheniformis DSM 17236 and Saccharomyces cerevisiae cell wall extract on dextran sulfate sodium (DSS)-induced colitis in Sprague Dawley rats. METHODS Acute colitis was induced in rats by oral administration of DSS 3.5% for 7 days. Fifty rats were divided equally into five groups; one control group and the other groups were induced with colitis and treated with or without the tested synbiotic, mixed with diet, for 28 days and sulfasalazine (100 mg/kg) via intragastric tube once daily for 14 days. RESULTS Symptomatically, the synbiotic administration raised the disease activity index (DAI) to comparable scores of the DSS group, specially from the 2nd to 7th days post DSS intoxication. It also induced a significant (p < 0.05) amplification of WBCs, myeloperoxidase (MPO), malondialdehyde (MDA), nuclear factor kappa B (NF-kB) expression and proinflammatory cytokines tumor necrosis factor alpha (TNFα), interferon gamma (INFγ), and interleukin-1 beta (IL-1β) while depressed the antioxidant enzymes glutathione peroxidase (GPx), catalase (CAT), and superoxide dismutase (SOD) when compared with the DSS and control groups. The DSS intoxicated and Synbiotic+DSS groups showed desquamations of the covering epithelium, noticeable diffuse leukocytic infiltrations, sever catarrhal enteritis, ischemic colitis with diffuse coagulative necrosis of the entire colonic mucosa. Contrarily, sulfasalazine proved to be effective in the reduction of the tested inflammatory markers and the pathological degenerative changes of the DSS ulcerative colitis. CONCLUSION The examined synbiotic did not ameliorate but aggravated the DSS-induced colitis, so it should be subjected to intensive experimental and clinical testing before their use in animals and human.
Collapse
Affiliation(s)
- Gamal A Salem
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Amany Abdel-Rahman Mohamed
- Departments of Forensic Medicine and Toxicology and Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Wael A M Ghonimi
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - H M Abdallah
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Nasreddin R Rhouma
- Department of Micobiology, Faculty of Science, Misurata University, Misurata, P.O. Box 2478, Libya
| | - Reem I Ali
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Banha University, Banha, 13518, Egypt
| |
Collapse
|
47
|
Rastogi S, Singh A. Gut microbiome and human health: Exploring how the probiotic genus Lactobacillus modulate immune responses. Front Pharmacol 2022; 13:1042189. [PMID: 36353491 PMCID: PMC9638459 DOI: 10.3389/fphar.2022.1042189] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
The highest density of microbes resides in human gastrointestinal tract, known as “Gut microbiome”. Of note, the members of the genus Lactobacillus that belong to phyla Firmicutes are the most important probiotic bacteria of the gut microbiome. These gut-residing Lactobacillus species not only communicate with each other but also with the gut epithelial lining to balance the gut barrier integrity, mucosal barrier defence and ameliorate the host immune responses. The human body suffers from several inflammatory diseases affecting the gut, lungs, heart, bone or neural tissues. Mounting evidence supports the significant role of Lactobacillus spp. and their components (such as metabolites, peptidoglycans, and/or surface proteins) in modulatingimmune responses, primarily through exchange of immunological signals between gastrointestinal tract and distant organs. This bidirectional crosstalk which is mediated by Lactobacillus spp. promotes anti-inflammatory response, thereby supporting the improvement of symptoms pertaining to asthma, chronic obstructive pulmonary disease (COPD), neuroinflammatory diseases (such as multiple sclerosis, alzheimer’s disease, parkinson’s disease), cardiovascular diseases, inflammatory bowel disease (IBD) and chronic infections in patients. The metabolic disorders, obesity and diabetes are characterized by a low-grade inflammation. Genus Lactobacillus alleviates metabolic disorders by regulating the oxidative stress response and inflammatory pathways. Osteoporosis is also associated with bone inflammation and resorption. The Lactobacillus spp. and their metabolites act as powerful immune cell controllers and exhibit a regulatory role in bone resorption and formation, supporting bone health. Thus, this review demonstrated the mechanisms and summarized the evidence of the benefit of Lactobacillus spp. in alleviating inflammatory diseases pertaining to different organs from animal and clinical trials. The present narrative review explores in detail the complex interactions between the gut-dwelling Lactobacillus spp. and the immune components in distant organs to promote host’s health.
Collapse
|
48
|
Tarapatzi G, Filidou E, Kandilogiannakis L, Spathakis M, Gaitanidou M, Arvanitidis K, Drygiannakis I, Valatas V, Kotzampassi K, Manolopoulos VG, Kolios G, Vradelis S. The Probiotic Strains Bifidοbacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii Regulate Wound Healing and Chemokine Responses in Human Intestinal Subepithelial Myofibroblasts. Pharmaceuticals (Basel) 2022; 15:1293. [PMID: 36297405 PMCID: PMC9611312 DOI: 10.3390/ph15101293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Bifidobacterium lactis, Lactobacillus acidophilus, Lactiplantibacillus plantarum and Saccharomyces boulardii are common probiotic supplements. Colonic subepithelial myofibroblasts (cSEMFs) are actively involved in mucosal wound healing and inflammation. cSEMFs, isolated from healthy individuals, were stimulated with 102 or 104 cfu/mL of these probiotic strains alone and in combination, and their effect on chemokine and wound healing factor expression was assessed by qRT-PCR, ELISA and Sircol Assay, and on cSEMFs migration, by Wound Healing Assay. These strains remained viable and altered cSEMFs’ inflammatory and wound healing behavior, depending on the strain and concentration. cSEMFs treated with a combination of the four probiotics had a moderate, but statistically significant, increase in the mRNA and/or protein expression of chemokines CXCL1, CXCL2, CXCL4, CXCL8, CXCL10, CCL2 and CCL5, and healing factors, collagen type I and III, fibronectin and tissue factor. In contrast, when each strain was administered alone, different effects were observed, with greater increase or decrease in chemokine and healing factor expression, which was balanced by the mixture. Overall, this study highlights that the use of multiple probiotic strains can potentially alert the gut mucosal immune system and promote wound healing, having a better effect on mucosal immunity than the use of single probiotics.
Collapse
Affiliation(s)
- Gesthimani Tarapatzi
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Michail Spathakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Maria Gaitanidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Ioannis Drygiannakis
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Valatas
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Katerina Kotzampassi
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Stergios Vradelis
- Second Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
49
|
Sabouri S, Esmailzadeh M, Sadeghinejad A, Eslami Shahrbabaki M, Asadikaram G, Nikvarz N. The Effect of Adjunctive Probiotics on Markers of Inflammation and Oxidative Stress in Bipolar Disorder: A Double-blind, Randomized, Controlled Trial. J Psychiatr Pract 2022; 28:373-382. [PMID: 36074106 DOI: 10.1097/pra.0000000000000660] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Bipolar disorder (BD) is a chronic psychiatric illness. Concentrations of inflammatory cytokines are increased in BD. Supplementation with probiotics has shown promising effects in reducing inflammation and producing improvement in clinical symptoms in some psychiatric disorders. Therefore, we designed a clinical trial to assess the effects of adjunctive probiotics on markers of inflammation and oxidative stress in patients with BD. METHODS In this 8-week, double-blind, randomized study, 38 patients suffering from BD type I were given a probiotic or placebo capsule each day. Serum levels of interleukin-6 (IL-6), as the primary outcome measure, and of interleukin-10 (IL-10), tumor necrosis factor-α, and malondialdehyde, as the secondary outcome measures, were obtained before and after the intervention. RESULTS At the end of the study, the 2 groups showed no significant or clinically meaningful differences in the serum concentrations of IL-6 [Hedge g=0.02, 95% confidence interval (CI): -0.6; 0.64, P=0.936], tumor necrosis factor-α (Hedge g=-0.2, 95% CI: -0.82; 0.42, P=0.554), IL-10 (Hedge g=-0.072, 95% CI: -0.071; 0.56, P=0.827), and malondialdehyde (Hedge g=0.27, 95% CI: -0.37; 0.91, P=0.423). CONCLUSIONS This study did not find any significant or conclusive effects of probiotics supplementation on markers of inflammation and oxidative stress in patients with BD. Further studies are needed before a conclusion can be drawn about the efficacy of probiotics in the management of BD.
Collapse
|
50
|
Xu L, Liu B, Huang L, Li Z, Cheng Y, Tian Y, Pan G, Li H, Xu Y, Wu W, Cui Z, Xie L. Probiotic Consortia and Their Metabolites Ameliorate the Symptoms of Inflammatory Bowel Diseases in a Colitis Mouse Model. Microbiol Spectr 2022; 10:e0065722. [PMID: 35730951 PMCID: PMC9430814 DOI: 10.1128/spectrum.00657-22] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022] Open
Abstract
Inflammatory bowel disease (IBD) has become a global public health problem. Although the pathogenesis of the disease is unknown, a potential association between the gut microbiota and inflammatory signatures has been established. Probiotics, especially Lactobacillus or Bifidobacterium, are orally taken as food supplements or microbial drugs by patients with IBD or gastrointestinal disorders due to their safety, efficacy, and power to restore the gut microenvironment. In the current study, we investigated the comprehensive effects of probiotic bacterial consortia consisting of Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus acidophilus (Lactobacillus spp.), and Bifidobacterium lactis (Bifidobacterium spp.) or their metabolites in a dextran sodium sulfate (DSS)-induced colitis mouse model. Our data demonstrate that probiotic consortia not only ameliorate the disease phenotype but also restore the composition and structure of the gut microbiota. Moreover, the effect of probiotic consortia is better than that of any single probiotic strain. The results also demonstrate that mixed fermentation metabolites are capable of ameliorating the symptoms of gut inflammation. However, the administration of metabolites is not as effective as probiotic consortia with respect to phenotypic characteristics, such as body weight, disease activity index (DAI), and histological score. In addition, mixed metabolites led only to changes in intestinal flora composition. In summary, probiotic consortia and metabolites could exert protective roles in the DSS-induced colitis mouse model by reducing inflammation and regulating microbial dysbiosis. These findings from the current study provide support for the development of probiotic-based microbial products as an alternative therapeutic strategy for IBD. IMPORTANCE IBD is a chronic nonspecific inflammatory disease. IBD is characterized by a wide range of lesions, often involving the entire colon, and is characterized mainly by ulcers and erosions of the colonic mucosa. In the present study, we investigated the efficacy of probiotics on the recovery of gut inflammation and the restoration of gut microecology. We demonstrate that probiotic consortia have a superior effect in inhibiting inflammation and accelerating recovery compared with the effects observed in the control group or groups administered with a single strain. These results support the utilization of probiotic consortia as an alternative therapeutic approach to treat IBD.
Collapse
Affiliation(s)
- Limin Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Bingdong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liujing Huang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ze Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yanbo Cheng
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ye Tian
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guihua Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Huijun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zongbin Cui
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liwei Xie
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|