1
|
Zaharuddin AM, Muslim A, Aazmi S, Idorus MY, Almabhouh FA, Lim SY, Loganathan AL, Ayub Q, Chong CW, Khalil KA, Ghani NA, Lim SM, Ramasamy K. Probiotic Lactobacillus rhamnosus GG Alleviates Prehypertension and Restores Gut Health and Microbiota in NaCl-Induced Prehypertensive Rats. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10536-z. [PMID: 40254701 DOI: 10.1007/s12602-025-10536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Probiotics could be used as adjuvant treatments in prehypertension management to restore gut microbiota dysbiosis caused by a high-salt diet. This study investigated the antihypertensive effects of the probiotic Lactobacillus rhamnosus strain GG (LGG) on high-salt diet-induced prehypertensive rats. Eighteen Sprague-Dawley rats were assigned equally into three groups: normotensive fed on a normal diet (ND), prehypertensive induced on a 4% NaCl high-salt diet (HSD), and prehypertensive induced on an HSD treated with LGG at 1 × 109 CFU daily for 8 weeks (LGG). Weekly changes in water, food, body weight, diastolic blood pressure (DBP), systolic blood pressure (SBP), and mean arterial pressure (MAP) were monitored. Serum levels of Na, K, Cl, ALB, Ca, and TP were measured at the end of treatment, along with morphological and histomorphometric changes in the small intestine. Stool samples collected before (W0) and 8 weeks after treatment (W8) were sequenced for bacterial 16S rDNA metagenomics. Probiotic LGG significantly reduces average DBP, SBP, and MAP while improving gut integrity through intact intestine morphology, higher villus heights, and a V/C ratio. At the genus level, the LGG group's gut microbiota composition is more similar to the HSD profile at W0 but shifts to the ND profile after treatment at W8. Thus, probiotic LGG lowers blood pressure indices, improves serum biochemistry profile, restores small intestinal integrity barrier, and modulates gut microbiota profile, indicating its potential as an adjuvant treatment for prehypertension and the significance of gut health in blood pressure regulation.
Collapse
Affiliation(s)
- Anis Munira Zaharuddin
- Microbiome Health and Environment (MiHeaRT) Research Interest Group, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Azdayanti Muslim
- Microbiome Health and Environment (MiHeaRT) Research Interest Group, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, Kampus Sungai Buloh, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia
- Institute for Biodiversity and Sustainable Development, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Shafiq Aazmi
- Microbiome Health and Environment (MiHeaRT) Research Interest Group, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia.
| | - Mohd Yusri Idorus
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
| | - Fayez A Almabhouh
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Kampus Sungai Buloh, Jalan Hospital, 47000, Sungai Buloh, Selangor, Malaysia
- Department of Biology and Biotechnology, Faculty of Science Islamic University of Gaza, Gaza Strip, Palestine
| | - Shu Yong Lim
- Monash University Malaysia Genomics Facility, Monash University Malaysia, 47500, Subang Jaya, Selangor, Malaysia
| | - Aswini Leela Loganathan
- Monash University Malaysia Genomics Facility, Monash University Malaysia, 47500, Subang Jaya, Selangor, Malaysia
| | - Qasim Ayub
- Monash University Malaysia Genomics Facility, Monash University Malaysia, 47500, Subang Jaya, Selangor, Malaysia
| | - Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, 47500, Subang Jaya, Selangor, Malaysia
| | - Khalilah Abdul Khalil
- Microbiome Health and Environment (MiHeaRT) Research Interest Group, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Nurunajah Ab Ghani
- Atta-Ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA, Cawangan Selangor, Kampus Puncak Alam, 42300, Bandar Puncak Alam, Selangor, Malaysia
| |
Collapse
|
2
|
罗 嘉, Sodnomjamts B, 高 雪, 陈 晶, 余 政, 熊 莎, 曹 虹. [ Akkermansia muciniphila gavage improves gut-brain interaction disorders in gp120 transgenic mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:554-565. [PMID: 40159970 PMCID: PMC11955896 DOI: 10.12122/j.issn.1673-4254.2025.03.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Indexed: 04/02/2025]
Abstract
OBJECTIVES To explore the effect of A. muciniphila gavage on intestinal microbiota and gut-brain interaction disorders (DGBIs) in gp120tg transgenic mouse models of HIV-associated neurocognitive disorder (HAND). METHODS Intestinal microbiota was detected by 16S rRNA gene sequencing in 6-, 9-, and 12-month-old wild-type (WT) mice and gp120tg transgenic mice. The 12-month-old WT and transgenic mice were divided into 2 groups for daily treatment with PBS or A.muciniphila gavage (2×108 CFU/mouse) for 6 weeks. After the treatment, immunohistochemistry, ELISA and qPCR were used to detect changes in colonic expression levels of glycosylated mucins, MBP and IL-1β, eosinophil infiltration, serum lipopolysaccharide (LPS) levels, and colonic expressions of occludin, ZO-1, IL-10, TNF-α and INF-γ mRNA. Morris water maze test and immunofluorescence assay were used to assess learning and spatial memory abilities and neuronal damage of the mice. RESULTS Compared with WT mice, the transgenic mice exhibited significantly lowered Simpson's diversity of the intestinal microbiota with reduced abundance of Akkermansia genus, increased serum LPS levels and decreased colonic expression of glycosylated mucin. A.muciniphila gavage obviously ameliorated the reduction of glycosylated mucin in the transgenic mice without causing significant changes in body weight. The 12-month-old gp120tg mice had significantly decreased cdonic expressions of Occludin and ZO-1 with increased eosinophil infiltration and TNF-β, INF-γ and IL-1β levels and obviously lowered IL-10 level; all these changes were significantly mitigated by A.muciniphila gavage, which also improved cognitive impairment and neuronal loss in the hippocampus and cortex of the transgenic mice. CONCLUSIONS The gp120tg mice have lower intestinal microbiota richness and diversity than WT mice. The 12-month-old gp120tg mice have significantly reduced Akkermansia abundance with distinct DGBIs-related indexes, and A. muciniphila gavage can reduce intestinal barrier injury, colonic inflammation and eosinophil activation, cognitive impairment and brain neuron injury in these mice.
Collapse
|
3
|
Wilson RM, Walker JM, Beld J, Yin K. Lactobacillus acidophilus (strain Scav) postbiotic metabolites reduce infection and modulate inflammation in an in vivo model of Pseudomonas aeruginosa wound infection. J Appl Microbiol 2025; 136:lxaf061. [PMID: 40068933 PMCID: PMC11951090 DOI: 10.1093/jambio/lxaf061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/30/2025]
Abstract
AIMS This study assessed the antibacterial, antibiofilm, and immunomodulatory activity of Lactobacillus acidophilus (strain Scav) postbiotic (LaP) in a mouse model of Pseudomonas aeruginosa wound infection and evaluated the bioactive components of the LaP. METHODS AND RESULTS LaP was tested for Pseudomonas aeruginosa clearance and immunomodulatory activity during wound infection. We show that LaP applied 1 h after infection reduced tissue bacterial burden within 24 h, and this reduction persisted for 5 days. Ciprofloxacin given once at the exact same time did not reduce bacteria load as compared to vehicle controls. LaP reduced plasma IL-6 and MCP-1 levels after 5 days. Wound tissue IL-6 and MCP-1 levels were increased in infected vehicle mice at 5 days, but tissues from LaP-treated mice were similar to sham controls. LaP increased tissue IL-10 (antiinflammatory cytokine) levels. Ciprofloxacin decreased plasma and tissue IL-6 compared to vehicle controls but did not affect MCP-1 or IL-10 levels. To elucidate antibacterial and antibiofilm metabolite(s) in LaP, fractionation followed by Ps. aeruginosa antagonistic activity assays were performed. This was followed by liquid chromatography coupled to mass spectrometry (LCMS) analysis. Our analyses identified a low molecular weight, polar molecule, which had both antibacterial and antibiofilm activity. CONCLUSIONS Lactobacillus acidophilus secretes an antibacterial and antibiofilm metabolite that reduced pathogen burden and resolved systemic inflammation in a Pseudomonas aeruginosa wound infection model.
Collapse
Affiliation(s)
- Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| | - Jean M Walker
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| | - Joris Beld
- Department of Microbiology and Immunology, College of Medicine, Drexel University, Philadelphia, PA 19104, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences, Stratford, NJ 08084, USA
| |
Collapse
|
4
|
Jang SB, Kim Y, Yeo HC, Kang GH, An BC, Ryu Y, Chung MJ, Cho SG. Probiotic-Derived P8 Protein: Promoting Proliferation and Migration in Stem Cells and Keratinocytes. Int J Stem Cells 2025; 18:87-98. [PMID: 39491493 PMCID: PMC11867908 DOI: 10.15283/ijsc24107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Probiotics exert various effects on the body and provide different health benefits. Previous reports have demonstrated that the P8 protein (P8), isolated from Lactobacillus rhamnosus, has anticancer properties. However, its efficacy in stem cells and normal cells has not been reported. In this study, the effect of P8 on cell proliferation and wound healing was evaluated, investigating its underlying mechanism. Based on scratch assay results, we demonstrated that P8 treatment significantly increases wound healing by activating the cell cycle and promoting stem cell stemness. Cellular mechanisms were further investigated by culturing stem cells in a medium containing Lactobacillus-derived P8 protein, revealing its promotion of cell proliferation and migration. Also, it is found that P8 enhances the expression of stemness markers, such as OCT4 and SOX2, along with activation of the mitogen-activated protein kinase (MAPK) signaling and Hippo pathways. These results indicate that P8 can promote cell growth by increasing stem cell proliferation, migration, and stemness in a manner associated with MAPK and Hippo signaling, which could contribute to the increased wound healing after P8 treatment. Furthermore, P8 could promote wound healing in keratinocytes by activating the MAPK signaling pathways. These results suggest that P8 might be a promising candidate to enhance stem cell culture efficiency by activating cell proliferation, and enhance therapeutic effects in skin diseases.
Collapse
Affiliation(s)
- Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Han Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | | | | | - Yongku Ryu
- R&D Center, Cell Biotech Co., Ltd., Gimpo, Korea
| | | | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| |
Collapse
|
5
|
Xia Y, Hu L, Ren K, Han X, Sun Y, Li D. Embryonic exposure to 6:2 fluorotelomer alcohol mediates autism spectrum disorder-like behavior by dysfunctional microbe-gut-brain axis in mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 484:136739. [PMID: 39637794 DOI: 10.1016/j.jhazmat.2024.136739] [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: 08/19/2024] [Revised: 10/29/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
6:2 fluorotelomer alcohol (6:2 FTOH) is considered an emerging contaminant as a substitute for perfluoroalkyl and polyfluoroalkyl substances. Autism spectrum disorder (ASD) is a highly heterogeneous childhood neurodevelopmental disorder, the prevalence of which has been significantly increasing globally, possibly due to rising exposure to environmental pollutants. Additionally, the microbe-gut-brain axis plays a crucial role in the development of ASD. The purpose of study was to investigate the impact of embryonic 6:2 FTOH exposure on neurological development in mice and explore the potential involvement of the microbe-gut-brain. Pregnant mice were orally administered 6:2 FTOH from gestation day 8.5 until delivery, and follow-up testing was performed on day 22 post-delivery. The findings revealed that embryonic exposure to 6:2 FTOH led to ASD-like symptoms, cortical neuron apoptosis, glial cell activation, and abnormal synapse formation in mice. Furthermore, impairment of colonic barrier function, inflammatory response, and dysbiosis in gut microbiota were observed. Interestingly, supplementation with Lactobacillus rhamnosus GG during embryonic development mitigated these adverse outcomes. This study enhances our understanding of how environmental pollutants can impact neurological development in children and provides valuable insights for clinical prevention, diagnosis, and treatment strategies for non-genetic ASD.
Collapse
Affiliation(s)
- Yunhui Xia
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Liehai Hu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Ke Ren
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yun Sun
- Genetic Medicine Center, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
6
|
Si W, Zhao X, Li R, Li Y, Ma C, Zhao X, Bugno J, Qin Y, Zhang J, Liu H, Wang L. Lactobacillus rhamnosus GG induces STING-dependent IL-10 in intestinal monocytes and alleviates inflammatory colitis in mice. J Clin Invest 2025; 135:e174910. [PMID: 39895628 PMCID: PMC11785918 DOI: 10.1172/jci174910] [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: 08/16/2023] [Accepted: 11/27/2024] [Indexed: 02/04/2025] Open
Abstract
Preclinical and clinical observations indicate that the probiotic Lactobacillus rhamnosus GG (LGG) can modulate colonic inflammation. However, the underlying mechanisms have not been explored in depth. Here, we demonstrate that oral administration of live LGG alleviated inflammatory colitis by increasing IL-10 expression in intestinal Ly6C+ monocytes. Mechanistically, LGG induced IL-10 production via the stimulator of IFN genes (STING)/TBK1/NF-κB (RELA) signaling pathway in intestinal Ly6C+ monocytes, enhancing their immune-suppressive function. Elevated IL-10 subsequently activated IL-10 signaling in Ly6C+ monocytes, resulting in an IL-10-based autocrine regulatory loop and inhibition of proinflammatory cytokine production. Furthermore, LGG shifted the gut microbial community and its metabolic functions, leading to intestinal immune responses against colitis. Fecal microbiota transplantation from LGG-colonized mice alleviated immune checkpoint blockade-associated colitis. Our findings highlight the importance of STING signaling in IL-10-dependent antiinflammatory immunity and establish an empirical basis for developing oral administration of live LGG as an efficient and safe therapeutic strategy against inflammatory colitis.
Collapse
Affiliation(s)
- Wei Si
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, Quebec, Canada
| | - Ruitong Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yaopeng Li
- Pritzker School of Molecular Engineering and
| | - Cui Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaohan Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois, USA
| | - Yuchang Qin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongwei Liu
- The Laboratory of Microbiome and Microecological Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Liangliang Wang
- The Laboratory of Microbiome and Microecological Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Horvath M, Yang R, Castaneda DC, Callender M, Aiken ES, Voigt AY, Caldwell R, Fachi J, Di Luccia B, Scholar Z, Yu P, Salner A, Colonna M, Palucka K, Oh J. Species- and strain-specific microbial modulation of interferon, innate immunity, and epithelial barrier in 2D air-liquid interface respiratory epithelial cultures. BMC Biol 2025; 23:28. [PMID: 39875977 PMCID: PMC11776145 DOI: 10.1186/s12915-025-02129-7] [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: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The microbiome regulates the respiratory epithelium's immunomodulatory functions. To explore how the microbiome's biodiversity affects microbe-epithelial interactions, we screened 58 phylogenetically diverse microbes for their transcriptomic effect on human primary bronchial air-liquid interface (ALI) cell cultures. RESULTS We found distinct species- and strain-level differences in host innate immunity and epithelial barrier response. Strikingly, we found that host interferon, an antiviral response, was one of the most variable host processes. This variability was not driven by microbial phylogenetic diversity, bioburden, nor by the microbe's ability to stimulate other innate immunity pathways. CONCLUSIONS Microbial colonization differentially stimulates host gene expression with variations observed across phylogenetically diverse microbes and across different strains of the same species. Our study provides a foundation for understanding how the respiratory microbiome's biodiversity affects epithelial, and particularly antiviral, innate immunity.
Collapse
Affiliation(s)
- Mian Horvath
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- UCONN Health (University of Connecticut), Farmington, CT, 06030, USA
| | - Ruoyu Yang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- UCONN Health (University of Connecticut), Farmington, CT, 06030, USA
- Duke University School of Medicine, Durham, NC, 27708, USA
| | | | - Megan Callender
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Elizabeth S Aiken
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Anita Y Voigt
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- Duke University School of Medicine, Durham, NC, 27708, USA
| | - Ryan Caldwell
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - José Fachi
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Blanda Di Luccia
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zoe Scholar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Peter Yu
- Hartford Healthcare Cancer Institute, Hartford, CT, 06102, USA
| | - Andrew Salner
- Hartford Healthcare Cancer Institute, Hartford, CT, 06102, USA
| | - Marco Colonna
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Julia Oh
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Duke University School of Medicine, Durham, NC, 27708, USA.
| |
Collapse
|
8
|
Zhao Y, Zhu S, Dong Y, Xie T, Chai Z, Gao X, Dai Y, Wang X. The Role of Gut Microbiome in Irritable Bowel Syndrome: Implications for Clinical Therapeutics. Biomolecules 2024; 14:1643. [PMID: 39766350 PMCID: PMC11674646 DOI: 10.3390/biom14121643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder (FGID) characterized by chronic or recurrent gastrointestinal symptoms without organic changes, and it is also a common disorder of gut-brain interaction (DGBIs).. The symptoms of IBS not only affect the quality of life for individual patients but also place a significant burden on global healthcare systems. The lack of established and universally applicable biomarkers for IBS, along with the substantial variability in symptoms and progression, presents challenges in developing effective clinical treatments. In recent years, preclinical and clinical studies have linked the pathogenesis of IBS to alterations in the composition and function of the intestinal microbiota. Within the complex microbial community of the gut, intricate metabolic and spatial interactions occur among its members and between microbes and their hosts. Amid the multifaceted pathophysiology of IBS, the role of intestinal microenvironment factors in symptom development has become more apparent. This review aims to delve into the changes in the composition and structure of the gut microbiome in individuals with IBS. It explores how diet-mediated alterations in intestinal microbes and their byproducts play a role in regulating the pathogenesis of IBS by influencing the "brain-gut" axis, intestinal barrier function, immune responses, and more. By doing so, this review seeks to lay a theoretical foundation for advancing the development of clinical therapeutics for IBS.
Collapse
Affiliation(s)
- Yucui Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shixiao Zhu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingling Dong
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tian Xie
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhiqiang Chai
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiumei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
| | - Yongna Dai
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
| | - Xiaoying Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
9
|
Meng Z, Huang X, Qiao M, Song L, Liu Y, Hai D. Lactic Acid Bacteria Surface Proteins in the Mechanisms of Cell Adhesion and Immunoregulation. Food Sci Nutr 2024; 12:10148-10163. [PMID: 39723039 PMCID: PMC11666997 DOI: 10.1002/fsn3.4517] [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: 01/30/2024] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 12/28/2024] Open
Abstract
This study delves into the role of lactic acid bacteria (LAB) surface proteins in cell adhesion and immunoregulation. Using fluorescence microscopy, we observed distinct adhesion patterns on various cell types. LAB surface proteins demonstrated concentration-dependent inhibition of Salmonella adhesion, with LAB69 exhibiting potent antagonistic effects. Genetic expression analysis revealed nuanced responses in key genes (MD2, TLR4, IL-10, MUC3, MIF) across different cell types, highlighting the diverse immunomodulatory effects of LAB surface proteins. Modulation of pro-inflammatory (TNF-α) and anti-inflammatory (IL-10) cytokines further emphasized the complex interplay. In conclusion, this study underscores the pivotal role of LAB surface proteins in mediating cell adhesion and immunoregulation, providing a foundation for isolating specific immunomodulatory molecules within LAB surface proteins for potential applications in microbial ecological agents.
Collapse
Affiliation(s)
- Ziheng Meng
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Xianqing Huang
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Mingwu Qiao
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Lianjun Song
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| | - Yufei Liu
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
| | - Dan Hai
- College of Food Science and TechnologyHenan Agricultural UniversityZhengzhouChina
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety ControlZhengzhouChina
| |
Collapse
|
10
|
Yi D, Liu X, Wang M, Zhao L, Liu Y, Xu Z, Peng Y, Zhang R, Wei Q, Liang Z, He J. Rosmarinic acid alleviated intestinal barrier damage caused by Escherichia coli by regulating the gut microbiota and inhibiting the NF-κB signalling pathway in mice. Food Funct 2024; 15:11740-11756. [PMID: 39540591 DOI: 10.1039/d4fo02654c] [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: 11/16/2024]
Abstract
Escherichia coli (E. coli) is a common zoonotic foodborne pathogen that poses a major threat to public health and economic development. Rosmarinic acid (RA) can inhibit intestinal inflammation; however, the protective effect of RA against the intestinal barrier damage induced by E. coli in mice and the underlying mechanism have not been elucidated. In this study, mice were orally administered with RA (20 mg kg-1) by gavage for one week and then were intraperitoneally challenged with E. coli. Mouse colonic epithelial cells (MCECs) were pretreated with RA for 6 h and challenged with E. coli (MOI = 1000) for 3 h. The results revealed that RA alleviated E. coli-induced weight loss in mice; reduced the increase in the levels of TNF-α, IL-6 and IL-1β in the serum; alleviated the decrease in ZO-1 protein expression; and increased intestinal permeability by inhibiting the NF-κB signalling pathway both in vivo and in vitro. Moreover, RA relieved the increase in intestinal permeability, reversed the structural damage to the mouse gut microbiota caused by E. coli, and increased the abundance of beneficial bacteria, including Lachnospiraceae_NK4136_group. Additionally, RA lost its protective function against E. coli infection in a pseudosterile mouse model, suggesting that the protection induced by RA was dependent on the gut microbiota. In conclusion, these results indicate that RA alleviates E. coli-induced inflammatory damage to the intestinal barrier by inhibiting the NF-κB signalling pathway and maintaining gut microbiota homeostasis. These findings provide new ideas and foundations for the application of RA as protection against E. coli.
Collapse
Affiliation(s)
- Dandan Yi
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Xia Liu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Menghui Wang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Linyi Zhao
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Yu Liu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Zhiran Xu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Ying Peng
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Rui Zhang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Qianyin Wei
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, PR China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, PR China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, PR China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, PR China
| |
Collapse
|
11
|
Zhang X, Sun L, Wu M, Yu C, Zhao D, Wang L, Zhang Z, Yi D, Hou Y, Wu T. Effect of supplementation with Lactobacillus rhamnosus GG powder on intestinal and liver damage in broiler chickens challenged by lipopolysaccharide. Front Microbiol 2024; 15:1466274. [PMID: 39534507 PMCID: PMC11555397 DOI: 10.3389/fmicb.2024.1466274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
This study explores the effect of dietary along with Lactobacillus rhamnosus GG (LGG) powder on intestinal and liver damage in broiler chickens challenged by lipopolysaccharide (LPS). A total of 100 healthy 1-day-old Ross 308 broiler chickens were selected and randomly divided into two treatments: the control group and the LGG treatment group. There were five replicates for each group, with 10 chickens per replicate. The chickens in the control group were fed a basal diet, while LGG treatment was supplemented with 1,000 mg/kg LGG along with the basal diet. The experiment lasted 29 days, and the trial included two phases. During the first 27 days, the animals were weighed on the 14th and 27th days to calculate growth performance. Then, on day 29, 2 animals from each replicate were intraperitoneally injected with 1 mg/kg BW LPS, and another 2 animals were treated with an equal volume of saline. The chickens were slaughtered 3 h later for sampling and further analysis. (1) LGG addition to the diet did not affect growth performance, including average daily gain (ADG), average daily feed intake (ADFI), and feed-to-weight ratio (F/G) of broiler chickens; (2) LPS stimulation decreased villus height (VH), and caused oxidative stress and increased the amount of diamine oxidase (DAO) in plasma, and the relative expression of intestinal inflammation genes (interleukin-8 [IL-8], interleukin 1β [IL-1β], inducible nitric oxide synthase [iNOS], and tumor necrosis factor-α [TNF-α]) and the relative expression of liver injury genes (b-cell lymphoma 2 [BCL2], heat shock protein70 [HSP70], and matrix metallopeptidase 13 [MMP13]). (3) Supplementation of LGG increased VH and the relative expression of intestinal barrier genes (mucins 2 [Mucin2] and occludin [Occludin]) and decreased the amount of DAO in plasma and the relative expression of intestinal inflammatory factors (IL-8, iNOS, and IL-1β). LGG supplementation also increased the expression of liver injury-related genes (MMP13 and MMP9). In conclusion, LGG enhanced intestinal barrier function, improved intestinal morphology, and alleviated the intestines' inflammatory response in LPS-stimulated broiler chicken, and it has a slightly protective effect on liver damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
12
|
Cheng X, Ren C, Mei X, Jiang Y, Zhou Y. Gut microbiota and irritable bowel syndrome: status and prospect. Front Med (Lausanne) 2024; 11:1429133. [PMID: 39484201 PMCID: PMC11524842 DOI: 10.3389/fmed.2024.1429133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Irritable bowel syndrome (IBS) is a very common gastrointestinal disease that, although not as aggressive as tumors, affects patients' quality of life in different ways. The cause of IBS is still unclear, but more and more studies have shown that the characteristics of the gut microbiota, such as diversity, abundance, and composition, are altered in patients with IBS, compared to the healthy population, which confirms that the gut microbiota plays a crucial role in the development of IBS. This paper aims to identify the commonalities by reviewing a large body of literature. Changes in the characteristics of gut microbiota in patients with different types of IBS are discussed, relevant mechanisms are described, and the treatment modalities of gut microbiota in IBS are summarized. Although there are more clinical trials that have made good progress, more standardized, more generalized, larger-scale, multi-omics clinical studies are what is missing. Overall, gut microbiota plays a crucial role in the development of IBS, and there is even more potential for treating IBS by modulating gut microbiota.
Collapse
Affiliation(s)
- Xinyu Cheng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Cheng Ren
- Department of Cardiology, The First people’s Hospital of Zhangjiagang, Affiliated Hospital of Soochow University, Medical Center of Soochow University, Zhangjiagang, Jiangsu, China
| | - Xiaofei Mei
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Pang X, Zhou B, Wu J, Mo Q, Yang L, Liu T, Jin G, Zhang L, Liu X, Xu X, Wang B, Cao H. Lacticaseibacillus rhamnosus GG alleviates sleep deprivation-induced intestinal barrier dysfunction and neuroinflammation in mice. Food Funct 2024; 15:8740-8758. [PMID: 39101469 DOI: 10.1039/d4fo00244j] [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: 08/06/2024]
Abstract
Consuming probiotic products is a solution that people are willing to choose to augment health. As a global health hazard, sleep deprivation (SD) can cause both physical and mental diseases. The present study investigated the protective effects of Lacticaseibacillus rhamnosus GG (LGG), a widely used probiotic, on a SD mouse model. Here, it has been shown that SD induced intestinal damage in mice, while LGG supplementation attenuated disruption of the intestinal barrier and enhanced the antioxidant capacity. Microbiome analysis revealed that SD caused dysbiosis in the gut microbiota, characterized by increased levels of Clostridium XlVa, Alistipes, and Desulfovibrio, as well as decreased levels of Ruminococcus, which were partially ameliorated by LGG. Moreover, SD resulted in elevated pro-inflammatory cytokine concentrations in both the intestine and the brain, while LGG provided protection in both organs. LGG supplementation significantly improved locomotor activity in SD mice. Although heat-killed LGG showed some protective effects in SD mice, its overall efficacy was inferior to that of live LGG. In terms of mechanism, it was found that AG1478, an inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase, could diminish the protective effects of LGG. In conclusion, LGG demonstrated the ability to alleviate SD-induced intestinal barrier dysfunction through EGFR activation and alleviate neuroinflammation.
Collapse
Affiliation(s)
- Xiaoqi Pang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Bingqian Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Jingyi Wu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Qi Mo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Lijiao Yang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Lan Zhang
- Department of Geriatrics, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Xin Xu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Anshan Road No. 154, Heping District, Tianjin, 300052, China.
| |
Collapse
|
14
|
He YQ, Zhu JR, Sun WJ, Luo YY, Wu XF, Yang M, Chen DF. ZO-1 and IL-1RAP Phosphorylation: Potential Role in Mediated Brain-Gut Axis Dysregulation in Irritable Bowel Syndrome-like Stressed Mice. Int J Med Sci 2024; 21:1738-1755. [PMID: 39006851 PMCID: PMC11241095 DOI: 10.7150/ijms.95848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/22/2024] [Indexed: 07/16/2024] Open
Abstract
Background and Objectives: Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder often exacerbated by stress, influencing the brain-gut axis (BGA). BGA dysregulation, disrupted intestinal barrier function, altered visceral sensitivity and immune imbalance defects underlying IBS pathogenesis have been emphasized in recent investigations. Phosphoproteomics reveals unique phosphorylation details resulting from environmental stress. Here, we employ phosphoproteomics to explore the molecular mechanisms underlying IBS-like symptoms, mainly focusing on the role of ZO-1 and IL-1RAP phosphorylation. Materials and Methods: Morris water maze (MWM) was used to evaluate memory function for single prolonged stress (SPS). To assess visceral hypersensitivity of IBS-like symptoms, use the Abdominal withdrawal reflex (AWR). Colonic bead expulsion and defecation were used to determine fecal characteristics of the IBS-like symptoms. Then, we applied a phosphoproteomic approach to BGA research to discover the molecular mechanisms underlying the process of visceral hypersensitivity in IBS-like mice following SPS. ZO-1, p-S179-ZO1, IL-1RAP, p-S566-IL-1RAP and GFAP levels in BGA were measured by western blotting, immunofluorescence staining, and enzyme-linked immunosorbent assay to validate phosphorylation quantification. Fluorescein isothiocyanate-dextran 4000 and electron-microscopy were performed to observe the structure and function of the intestinal epithelial barrier. Results: The SPS group showed changes in learning and memory ability. SPS exposure affects visceral hypersensitivity, increased fecal water content, and significant diarrheal symptoms. Phosphoproteomic analysis displayed that p-S179-ZO1 and p-S566-IL-1RAP were significantly differentially expressed following SPS. In addition, p-S179-ZO1 was reduced in mice's DRG, colon, small intestine, spinal and hippocampus and intestinal epithelial permeability was increased. GFAP, IL-1β and p-S566-IL-1RAP were also increased at the same levels in the BGA. And IL-1β showed no significant difference was observed in serum. Our findings reveal substantial alterations in ZO-1 and IL-1RAP phosphorylation, correlating with increased epithelial permeability and immune imbalance. Conclusions: Overall, decreased p-S179-ZO1 and increased p-S566-IL-1RAP on the BGA result in changes to tight junction structure, compromising the structure and function of the intestinal epithelial barrier and exacerbating immune imbalance in IBS-like stressed mice.
Collapse
Affiliation(s)
- Yu-Qin He
- Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jian-Ru Zhu
- Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Wen-Jing Sun
- Department of Gastroenterology and Hepatology, The Thirteenth People's Hospital of Chongqing, 400030, China
| | - Yuan-Yuan Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Xiao-Feng Wu
- Department of Stem Cell and Regenerative Medicine, Gastroenterology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Min Yang
- Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Dong-Feng Chen
- Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| |
Collapse
|
15
|
Zhu R, Zhao X, Wu H, Zeng X, Wei J, Chen T. Psychobiotics Lactiplantibacillus plantarum JYLP-326: Antidepressant-like effects on CUMS-induced depressed mouse model and alleviation of gut microbiota dysbiosis. J Affect Disord 2024; 354:752-764. [PMID: 38537753 DOI: 10.1016/j.jad.2024.03.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Depression affects a significant portion of the global population and has emerged as one of the most debilitating conditions worldwide. Recent studies have explored the relationship between depression and the microbiota of the intestine, revealing potential avenues for effective treatment. METHODS To evaluate the potential alleviation of depression symptoms, we employed a depression C57BL/6 mice model induced by chronic unpredictable mild stress (CUMS). We administered Lactiplantibacillus plantarum JYLP-326 and conducted various animal behavior tests, including the open-field test (OFT), sucrose preference test (SPT), and tail-suspension test (TST). Additionally, we conducted immunohistochemistry staining and analyzed the hippocampal and colon parts of the mice. RESULTS The results of the behavior tests indicated that L. plantarum JYLP-326 alleviated spontaneous behavior associated with depression. Moreover, the treatment led to significant improvements in GFAP and Iba1, suggesting its potential neuroprotective effects. Analysis of the hippocampal region indicated that L. plantarum JYLP-326 administration upregulated p-TPH2, TPH2, and 5-HT1AR, while downregulating the expression of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. In the colon, the treatment inhibited the TLR4-MyD88-NF-κB pathway and increased the levels of occludin and ZO-1, indicating improved intestinal barrier function. Additionally, the probiotic demonstrated a regulatory effect on the HMGB1-RAGE-TLR4 signaling pathway. CONCLUSIONS Our findings demonstrate that L. plantarum JYLP-326 exhibits significant antidepressant-like effects in mice, suggesting its potential as a therapeutic approach for depression through the modulation of gut microbiota. However, further investigations and clinical trials are required to validate its safety and efficacy for human use.
Collapse
Affiliation(s)
- Ruizhe Zhu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuanqi Zhao
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Heng Wu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiangdi Zeng
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China; School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
16
|
Wang J, Xue X, Zhao X, Luo L, Liu J, Dai S, Zhang F, Wu R, Liu Y, Peng C, Li Y. Forsythiaside A alleviates acute lung injury by inhibiting inflammation and epithelial barrier damages in lung and colon through PPAR-γ/RXR-α complex. J Adv Res 2024; 60:183-200. [PMID: 37579917 PMCID: PMC11156707 DOI: 10.1016/j.jare.2023.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/04/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023] Open
Abstract
INTRODUCTION Acute lung injury (ALI) is a lung disease characterized by inflammation and still requires further drug development. Forsythiaside A as the active compound of Forsythiae Fructus has the therapeutic potential for ALI. OBJECTIVE To investigate the mechanism of forsythiaside A in treating ALI through PPAR-γ and its conjugate RXR-α based on gut-lung axis. METHODS This study constructed in vitro and in vivo injury models using LPS and TNF-α. Forsythiaside A was used for the drug treatment, and RXR-α inhibitor UVI3003 was used to interfere with PPAR-γ/RXR-α complexes in the cells. HE staining was used for histopathological examination. Serum endotoxin contents were determined using limulus lysate kit. IHC staining and Western blot were conducted to assess the protein expressions. ELISA was applied to examine the content of pro-inflammatory cytokines in the cell supernatants. The protein interactions were analyzed via CO-IP. RESULTS In vivo results showed that forsythiaside A regulated PPAR-γ/RXR-α and inhibited TLR4/MAPK/NF-κB and MLCK/MLC2 signal pathways, thus inhibiting inflammation and epithelial barrier damages of lung and colon in ALI mice induced by intratracheal LPS. PPAR-γ/RXR-α were promoted by forsythiaside A in lungs, whereas inhibited by forsythiaside A in colons. Additionally, in vitro results showed that forsythiaside A suppressed inflammation and epithelial barrier damages in macrophages and lung/colon epithelial cells, by manipulating PPAR-γ/RXR-α to suppress the LPS- and TNF-α-induced activation of TLR4/MAPK/NF-κB and NF-κB/MLCK/MLC2 signal pathways. Moreover, further mechanism study indicated that forsythiaside A showed a cell-specific regulatory effect on PPAR-γ/RXR-α complex. Specifically, the PPAR-γ/RXR-α protein interactions were promoted by forsythiaside A in LPS-induced macrophages RAW264.7 and TNF-α-induced lung epithelial cells A549, but inhibited by forsythiaside A in TNF-α-induced colon epithelial cells SW620. CONCLUSION In the treatment of ALI, Forsythiaside A inhibited inflammation and epithelial barrier damages of lung and colon through its regulation on PPAR-γ/RXR-α complex.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lin Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Juan Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rui Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanfang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
17
|
Leser T, Baker A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG ® Probiotic Function. Microorganisms 2024; 12:794. [PMID: 38674738 PMCID: PMC11051730 DOI: 10.3390/microorganisms12040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
To advance probiotic research, a comprehensive understanding of bacterial interactions with human physiology at the molecular and cellular levels is fundamental. Lacticaseibacillus rhamnosus LGG® is a bacterial strain that has long been recognized for its beneficial effects on human health. Probiotic effector molecules derived from LGG®, including secreted proteins, surface-anchored proteins, polysaccharides, and lipoteichoic acids, which interact with host physiological processes have been identified. In vitro and animal studies have revealed that specific LGG® effector molecules stimulate epithelial cell survival, preserve intestinal barrier integrity, reduce oxidative stress, mitigate excessive mucosal inflammation, enhance IgA secretion, and provide long-term protection through epigenetic imprinting. Pili on the cell surface of LGG® promote adhesion to the intestinal mucosa and ensure close contact to host cells. Extracellular vesicles produced by LGG® recapitulate many of these effects through their cargo of effector molecules. Collectively, the effector molecules of LGG® exert a significant influence on both the gut mucosa and immune system, which promotes intestinal homeostasis and immune tolerance.
Collapse
Affiliation(s)
- Thomas Leser
- Future Labs, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark;
| | | |
Collapse
|
18
|
Abrehame S, Hung MY, Chen YY, Liu YT, Chen YT, Liu FC, Lin YC, Chen YP. Selection of Fermentation Supernatant from Probiotic Strains Exhibiting Intestinal Epithelial Barrier Protective Ability and Evaluation of Their Effects on Colitis Mouse and Weaned Piglet Models. Nutrients 2024; 16:1138. [PMID: 38674829 PMCID: PMC11053620 DOI: 10.3390/nu16081138] [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: 03/07/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The intestinal epithelial barrier can prevent the invasion of pathogenic microorganisms and food antigens to maintain a consistent intestinal homeostasis. However, an imbalance in this barrier can result in various diseases, such as inflammatory bowel disease, malnutrition, and metabolic disease. Thus, the aim of this study was to select probiotic strains with epithelial barrier-enhancing ability in cell-based model and further investigate them for their improving effects on colitis mouse and weaned piglet models. The results showed that selected specific cell-free fermentation supernatants (CFSs) from Ligilactobacillus salivarius P1, Lactobacillus gasseri P12, and Limosilactobacillus reuteri G7 promoted intestinal epithelial cell growth and proliferation, strengthening the intestinal barrier in an intestinal epithelial cell line Caco-2 model. Further, the administration of CFSs of L. salivarius P1, L. gasseri P12, and L. reuteri G7 were found to ameliorate DSS-induced colitis in mice. Additionally, spray-dried powders of CFS from the three strains were examined in a weaned piglet model, only CFS powder of L. reuteri G7 could ameliorate the feed/gain ratio and serum levels of D-lactate and endotoxin. In conclusion, a new potential probiotic strain, L. reuteri G7, was selected and showed ameliorating effects in both colitis mouse and weaned piglet models.
Collapse
Affiliation(s)
- Solomon Abrehame
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- Ethiopian Agricultural Authority, Ministry of Agriculture of Ethiopia (MoA), P.O. Box 62347, Addis Ababa 1000, Ethiopia
| | - Man-Yun Hung
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yu-Yi Chen
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yu-Tse Liu
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yung-Tsung Chen
- Department of Food Science, National Taiwan Ocean University, 2 Beining Road, Zhongzheng District, Keelung City 202, Taiwan
| | - Fang-Chueh Liu
- Animal Nutrition Division, Taiwan Livestock Research Institute, Ministry of Agriculture, 112 Farm Road, HsinHua District, Tainan City 712, Taiwan
| | - Yu-Chun Lin
- Animal Nutrition Division, Taiwan Livestock Research Institute, Ministry of Agriculture, 112 Farm Road, HsinHua District, Tainan City 712, Taiwan
- Fisheries Research Institute, Ministry of Agriculture, 199 Hou-Ih Road, Keelung City 202, Taiwan
| | - Yen-Po Chen
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| |
Collapse
|
19
|
Zheng J, Ahmad AA, Yang C, Liang Z, Shen W, Liu J, Yan Z, Han J, Yang Y, Dong P, Lan X, Salekdeh GH, Ding X. Orally Administered Lactobacillus rhamnosus CY12 Alleviates DSS-Induced Colitis in Mice by Restoring the Intestinal Barrier and Inhibiting the TLR4-MyD88-NF-κB Pathway via Intestinal Microbiota Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38598717 DOI: 10.1021/acs.jafc.3c07279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Oral ingestion of probiotics is a promising approach to relieving inflammatory disease through regulating the gut microbiota. A newly discovered strain, Lactobacillus rhamnosus CY12 (LCY12), obtained from cattle-yak milk, displayed numerous probiotic properties. These included enhanced viability in low pH and bile environments, adhesion capabilities, and potent antimicrobial effects. The research aimed to explore the beneficial impacts of the novel LCY12 strain on colitis in mice induced by dextran sulfate sodium (DSS) and to elucidate the underlying molecular mechanisms. The results of the study showed that administration of LCY12 effectively helped to reduce the negative effects of DSS-induced body weight loss, disease activity index score, colon length shortening, loss of goblet cells, and overall histopathological scores in the intestines. Simultaneously, LCY12 administration significantly alleviated intestinal inflammation and safeguarded intestinal barrier integrity by enhancing IL-10 levels, while dampening IL-6, IL-1β, and TNF-α production. Additionally, LCY12 boosted the presence of tight junction proteins. Furthermore, LCY12 hindered the TLR4/MyD88/NF-κB signaling pathway by downregulating TLR4 and MyD88 expression, inactivating phosphorylated IκBα, and preventing translocation of NF-κB p65 from the cytoplasm to the nucleus. The LCY12 also increased specific intestinal microbial communities and short-chain fatty acid (SCFA) production. Altogether, LCY12 oral administration alleviated colitis induced with DSS in mice by improving intestinal barrier function and regulating inflammatory cytokines, SCFA production, and intestinal microbiota.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Anum Ali Ahmad
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH8 9YL, U.K
| | - Chen Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zeyi Liang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jing Liu
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zuoting Yan
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jianlin Han
- Livestock Genetics Program, International Livestock Research Institute (ILRI), 00100 Nairobi, Kenya
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Pengcheng Dong
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs & Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| |
Collapse
|
20
|
Liu C, Qi X, Liu X, Sun Y, Mao K, Shen G, Ma Y, Li Q. Anti-inflammatory probiotics HF05 and HF06 synergistically alleviate ulcerative colitis and secondary liver injury. Food Funct 2024; 15:3765-3777. [PMID: 38506656 DOI: 10.1039/d3fo04419j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Given the limited efficacy and adverse effects associated with conventional drugs, probiotics are emerging as a promising therapeutic strategy for mitigating the chronic nature of ulcerative colitis (UC) and its consequential secondary liver injury (SLI). Limosilactobacillus fermentum HF06 and Lactiplatibacillus plantarum HF05 are strains we screened with excellent anti-inflammatory and probiotic properties in vitro. In this study, the intervention of HF06 and HF05 in combination (MIXL) was found to be more effective in alleviating intestinal inflammation and secondary liver injury in UC mice compared to supplementing with the two strains individually. Results demonstrated that MIXL effectively attenuated colon shortening and weight loss, downregulated the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 mRNA in the intestines, mitigated SLI, and augmented the enzymatic activities of SOD, CAT, and GSH-Px in the liver. MIXL enhances the intestinal barrier in UC mice, regulates the structure and composition of the gut microbiota, promotes the abundance of Lactobacillus, and suppresses the abundance of bacteria associated with inflammation and liver injury, including Clostridium_Sensu_Stricto_1, Escherichia, Shigella, Enterococcus, Corynebacterium, Desulfovibrio, and norank_f__Oscillospiraceae. This study demonstrated the synergistic effect of HF06 and HF05, providing a reliable foundation for the alleviation of UC.
Collapse
Affiliation(s)
| | - Xiaofen Qi
- Harbin Institute of Technology, Harbin, China.
| | - Xiaolin Liu
- Harbin Institute of Technology, Harbin, China.
| | - Yue Sun
- Harbin Institute of Technology, Harbin, China.
| | - Kaidong Mao
- Jiangsu HOWYOU Biotechnology Co., Ltd, Qidong, China
| | - Guiqi Shen
- Jiangsu HOWYOU Biotechnology Co., Ltd, Qidong, China
| | - Ying Ma
- Harbin Institute of Technology, Harbin, China.
| | - Qingming Li
- New Hope Dairy Company Limited, China.
- Sichuan Engineering Laboratory for High-quality Dairy Product Preparation and Quality Control Technology, China
| |
Collapse
|
21
|
Wu Y, Zhang X, Liu X, Li Y, Han D, Pi Y, Whitmore MA, Lu X, Zhang G, Zheng J, Wang J. Strain specificity of lactobacilli with promoted colonization by galactooligosaccharides administration in protecting intestinal barriers during Salmonella infection. J Adv Res 2024; 56:1-14. [PMID: 36894120 PMCID: PMC10834803 DOI: 10.1016/j.jare.2023.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
INTRODUCTION Galactooligosaccharides (GOS) are lactogenic prebiotics that exert health benefits by stimulating the growth of different Lactobacillus strains in the gastrointestinal (GI) tract. OBJECTIVES This study aimed to investigate the mechanism of action of different GOS-enriched lactobacilli in intestinal health. METHODS Piglets and mice were supplemented with GOS to identify specific enrichment of Lactobacillus. The protective effects of individual GOS-enriched lactobacilli were investigated in Salmonella-infected mice. Macrophage depletion and transcriptome analysis were further performed to assess the involvement of macrophages and the underlying mechanisms of individual lactobacilli. An in vitro cell co-culture system was also used to evaluate the anti-adhesive and anti-invasive activities of lactobacilli against Salmonella in epithelial cells. RESULTS GOS markedly increased the relative abundance of three lactobacilli including L. delbrueckii, L. johnsonii, and L. reuteri in both piglets and mice. Supplementation with GOS further alleviated Salmonella infection in mice. L. delbrueckii (ATCC®BAA 365™), but not L. johnsonii or L. reuteri, enhanced propionate production in the intestinal tract and ameliorated Salmonella-induced intestinal inflammation and barrier dysfunction by suppressing the JAK2-STAT3 signaling and M1 macrophage polarization. L. johnsonii (BNCC 186110), on the other hand, inhibited Salmonella adhesion and invasion of epithelial cells through competitive exclusion. However, L. reuteri (BNCC 186135) failed to protect mice against Salmonella infection. CONCLUSION GOS-enriched lactobacilli show a differential role in protecting against Salmonella-induced intestinal barrier dysfunction and inflammation. Our results provide novel insights into the mechanism of action of GOS and individual Lactobacillus strains in the control and prevention of intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoyi Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yi Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yu Pi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Melanie A Whitmore
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Xingmiao Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jinkai Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
22
|
Wiefels MD, Furar E, Eshraghi RS, Mittal J, Memis I, Moosa M, Mittal R, Eshraghi AA. Targeting Gut Dysbiosis and Microbiome Metabolites for the Development of Therapeutic Modalities for Neurological Disorders. Curr Neuropharmacol 2024; 22:123-139. [PMID: 36200211 PMCID: PMC10716879 DOI: 10.2174/1570159x20666221003085508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/10/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
The gut microbiota, composed of numerous species of microbes, works in synergy with the various organ systems in the body to bolster our overall health and well-being. The most well-known function of the gut microbiome is to facilitate the metabolism and absorption of crucial nutrients, such as complex carbohydrates, while also generating vitamins. In addition, the gut microbiome plays a crucial role in regulating the functioning of the central nervous system (CNS). Host genetics, including specific genes and single nucleotide polymorphisms (SNPs), have been implicated in the pathophysiology of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), and autism spectrum disorder (ASD). The gut microbiome dysbiosis also plays a role in the pathogenesis of these neurodegenerative disorders, thus perturbing the gut-brain axis. Overproduction of certain metabolites synthesized by the gut microbiome, such as short-chain fatty acids (SCFAs) and p-cresyl sulfate, are known to interfere with microglial function and trigger misfolding of alpha-synuclein protein, which can build up inside neurons and cause damage. By determining the association of the gut microbiome and its metabolites with various diseases, such as neurological disorders, future research will pave the way for the development of effective preventive and treatment modalities.
Collapse
Affiliation(s)
- Matthew D Wiefels
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Emily Furar
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rebecca S Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jeenu Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Idil Memis
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Moeed Moosa
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Rahul Mittal
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Adrien A Eshraghi
- Hearing Research and Communication Disorders Laboratory, Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
23
|
Cufaro MC, Prete R, Di Marco F, Sabatini G, Corsetti A, Gonzalez NG, Del Boccio P, Battista N. A proteomic insight reveals the role of food-associated Lactiplantibacillus plantarum C9O4 in reverting intestinal inflammation. iScience 2023; 26:108481. [PMID: 38213792 PMCID: PMC10783612 DOI: 10.1016/j.isci.2023.108481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/06/2023] [Accepted: 11/15/2023] [Indexed: 01/13/2024] Open
Abstract
Nowadays, Western diets and lifestyle lead to an increasing occurrence of chronic gut inflammation that represents an emerging health concern with still a lack of successful therapies. Fermented foods, and their associated lactic acid bacteria, have recently regained popularity for their probiotic potential including the maintenance of gut homeostasis by modulating the immune and inflammatory response. Our study aims to investigate the crosstalk between the food-borne strain Lactiplantibacillus plantarum C9O4 and intestinal epithelial cells in an in vitro inflammation model. Cytokines profile shows the ability of C9O4 to significantly reduce levels of IL-2, IL-5, IL-6, and IFN-γ. Proteomic functional analysis reveals an immunoregulatory role of C9O4, able to revert the detrimental effects of IFN-γ through the JAK/STAT pathway in inflamed intestinal cells. These results suggest a promising therapeutic role of fermented food-associated microbes for the management of gastrointestinal inflammatory diseases. Data are available via ProteomeXchange with identifier PXD042175.
Collapse
Affiliation(s)
- Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Roberta Prete
- Department of Bioscience for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Federica Di Marco
- Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giusi Sabatini
- Department of Bioscience for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Aldo Corsetti
- Department of Bioscience for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Natalia Garcia Gonzalez
- Department of Bioscience for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Natalia Battista
- Department of Bioscience for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
24
|
Agbemavor WSK, Buys EM. Dynamic Interactions between Diarrhoeagenic Enteroaggregative Escherichia coli and Presumptive Probiotic Bacteria: Implications for Gastrointestinal Health. Microorganisms 2023; 11:2942. [PMID: 38138086 PMCID: PMC10745617 DOI: 10.3390/microorganisms11122942] [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: 10/29/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
This study delves into the temporal dynamics of bacterial interactions in the gastrointestinal tract, focusing on how probiotic strains and pathogenic bacteria influence each other and human health. This research explores adhesion, competitive exclusion, displacement, and inhibition of selected diarrhoeagenic Escherichia coli (D-EAEC) and potential probiotic strains under various conditions. Key findings reveal that adhesion is time-dependent, with both D-EAEC K2 and probiotic L. plantarum FS2 showing increased adhesion over time. Surprisingly, L. plantarum FS2 outperformed D-EAEC K2 in adhesion and exhibited competitive exclusion and displacement, with inhibition of adhesion surpassing competitive exclusion. This highlights probiotics' potential to slow pathogen attachment when not in competition. Pre-infecting with L. plantarum FS2 before pathogenic infection effectively inhibited adhesion, indicating probiotics' ability to prevent pathogen attachment. Additionally, adhesion correlated strongly with interleukin-8 (IL-8) secretion, linking it to the host's inflammatory response. Conversely, IL-8 secretion negatively correlated with trans-epithelial electrical resistance (TEER), suggesting a connection between tight junction disruption and increased inflammation. These insights offer valuable knowledge about the temporal dynamics of gut bacteria interactions and highlight probiotics' potential in competitive exclusion and inhibiting pathogenic bacteria, contributing to strategies for maintaining gastrointestinal health and preventing infections.
Collapse
Affiliation(s)
- Wisdom Selorm Kofi Agbemavor
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Pretoria 0028, South Africa
- Radiation Technology Centre, Biotechnology and Nuclear Agriculture Research Institute, Ghana Atomic Energy Commission, Legon, Accra P.O. Box LG 80, Ghana
| | - Elna Maria Buys
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Pretoria 0028, South Africa
| |
Collapse
|
25
|
Song Q, Zhang X, Liu W, Wei H, Liang W, Zhou Y, Ding Y, Ji F, Ho-Kwan Cheung A, Wong N, Yu J. Bifidobacterium pseudolongum-generated acetate suppresses non-alcoholic fatty liver disease-associated hepatocellular carcinoma. J Hepatol 2023; 79:1352-1365. [PMID: 37459922 DOI: 10.1016/j.jhep.2023.07.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 06/07/2023] [Accepted: 07/04/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND & AIMS Recent studies have highlighted the role of the gut microbiota and their metabolites in non-alcoholic fatty liver disease-associated hepatocellular carcinoma (NAFLD-HCC). We aimed to identify specific beneficial bacterial species that could be used prophylactically to prevent NAFLD-HCC. METHODS The role of Bifidobacterium pseudolongum was assessed in two mouse models of NAFLD-HCC: diethylnitrosamine + a high-fat/high-cholesterol diet or + a choline-deficient/high-fat diet. Germ-free mice were used for the metabolic study of B. pseudolongum. Stool, portal vein and liver tissues were collected from mice for non-targeted and targeted metabolomic profiles. Two human NAFLD-HCC cell lines (HKCI2 and HKCI10) were co-cultured with B. pseudolongum-conditioned media (B.p CM) or candidate metabolites. RESULTS B. pseudolongum was the top depleted bacterium in mice with NAFLD-HCC. Oral gavage of B. pseudolongum significantly suppressed NAFLD-HCC formation in two mouse models (p < 0.01). Incubation of NAFLD-HCC cells with B.p CM significantly suppressed cell proliferation, inhibited the G1/S phase transition and induced apoptosis. Acetate was identified as the critical metabolite generated from B. pseudolongum in B.p CM, an observation that was confirmed in germ-free mice. Acetate inhibited cell proliferation and induced cell apoptosis in NAFLD-HCC cell lines and suppressed NAFLD-HCC tumor formation in vivo. B. pseudolongum restored heathy gut microbiome composition and improved gut barrier function. Mechanistically, B. pseudolongum-generated acetate reached the liver via the portal vein and bound to GPR43 (G coupled-protein receptor 43) on hepatocytes. GPR43 activation suppressed the IL-6/JAK1/STAT3 signaling pathway, thereby preventing NAFLD-HCC progression. CONCLUSIONS B. pseudolongum protected against NAFLD-HCC by secreting the anti-tumor metabolite acetate, which reached the liver via the portal vein. B. pseudolongum holds potential as a probiotic for the prevention of NAFLD-HCC. IMPACT AND IMPLICATIONS Non-alcoholic fatty liver disease-associated hepatocellular carcinoma (NAFLD-HCC) is an increasing healthcare burden worldwide. There is an urgent need to develop effective agents to prevent NAFLD-HCC progression. Herein, we show that the probiotic Bifidobacterium pseudolongum significantly suppressed NAFLD-HCC progression by secreting acetate, which bound to hepatic GPR43 (G coupled-protein receptor 43) via the gut-liver axis and suppressed the oncogenic IL-6/JAK1/STAT3 signaling pathway. Bifidobacterium pseudolongum holds potential as a novel probiotic for NAFLD-HCC prevention.
Collapse
Affiliation(s)
- Qian Song
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Weixin Liu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China; Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wei Liang
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yunfei Zhou
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Yanqiang Ding
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Fenfen Ji
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nathalie Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Hong Kong SAR, China.
| |
Collapse
|
26
|
Cui Y, Huang P, Duan H, Song S, Gan L, Liu Z, Lin Q, Wang J, Qi G, Guan J. Role of microencapsulated Lactobacillus plantarum in alleviating intestinal inflammatory damage through promoting epithelial proliferation and differentiation in layer chicks. Front Microbiol 2023; 14:1287899. [PMID: 38053557 PMCID: PMC10694250 DOI: 10.3389/fmicb.2023.1287899] [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: 09/03/2023] [Accepted: 10/23/2023] [Indexed: 12/07/2023] Open
Abstract
The alleviating effects of Lactobacillus plantarum in microencapsulation (LPM) on lipopolysaccharide (LPS)-induced intestinal inflammatory injury were investigated in layer chicks. A total of 252 healthy Hy-Line Brown layer chicks were randomly divided into six groups. Birds were injected with saline or LPS except for the control, and the diets of birds subjected to LPS were supplemented with nothing, L. plantarum, LPM, and wall material of LPM, respectively. The viable counts of LPM reached 109 CFU/g, and the supplemental levels of L. plantarum, LPM, and WM were 0.02 g (109 CFU), 1.0 g, and 0.98 g, per kilogram feed, respectively. LPS administration caused intestinal damage in layer chicks, evidenced by increased proinflammatory factors accompanied by poor intestinal development and morphology (p < 0.05). LPM/LPS significantly increased body weight, small intestine weight and length, villus height, villus height/crypt depth, and mRNA relative expression of tight junction protein genes (p < 0.05) and performed better than free L. plantarum. These findings could be attributed to the significant increase in viable counts of L. plantarum in the small intestine (p < 0.05), as well as the enhanced levels of Actinobacteriota, Lactobacillaceae, and Lactobacillus in intestinal microbiota (p < 0.05). Such results could further significantly increase goblet and PCNA+ cell percentage (p < 0.05); the mRNA relative expressions of epithelial cell, fast-cycling stem cell, quiescent stem cell, endocrine cell, and Paneth cell; and goblet and proliferative cell marker genes, including E-cadherin, Lgr-5, Bmi-1, ChA, Lysozome, Mucin-2, and PCNA (p < 0.05). Furthermore, the mRNA relative expressions of key genes involved in epithelial cell proliferation, namely, c-Myc, Cyclin-1, Wnt-3, Lrp-5, and Olfm-4, exhibited significant upregulation compared with the LPS treatment, as well as the differentiating genes Notch-1 and Hes-1 (p < 0.05). To sum up, microencapsulated L. plantarum supplementation could alleviate intestinal injury in layer chicks induced by LPS by promoting the proliferation and differentiation of intestinal epithelial cells, which could be attributed to the increase in viable count of L. plantarum in the gut and optimization in intestinal microbial flora.
Collapse
Affiliation(s)
- Yaoming Cui
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Peiyu Huang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Haitao Duan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| | - Shijia Song
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Liping Gan
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Zhen Liu
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Qiaohan Lin
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Jinrong Wang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| | - Gunghai Qi
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junjun Guan
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Zhang D, Zhou X, Zhou W, Cui SW, Nie S. Intestinal organoids: A thriving and powerful tool for investigating dietary nutrients-intestinal homeostasis axis. Food Res Int 2023; 172:113109. [PMID: 37689878 DOI: 10.1016/j.foodres.2023.113109] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 09/11/2023]
Abstract
Dietary nutrients regulate intestinal homeostasis through a variety of complex mechanisms, to affect the host health. Nowadays, various models have been used to investigate the dietary nutrients-intestinal homeostasis axis. Different from the limited flux in animal experiments, limited intestinal cell types and distorted simulation of intestinal environment of 2D cells, intestinal organoid (IO) is a 3D culture system of mini-gut with various intestinal epithelial cells (IECs) and producibility of intestinal biology. Therefore, IOs is a powerful tool to evaluate dietary nutrients-intestinal homeostasis interaction. This review summarized the application of IOs in the investigation of mechanisms for macronutrients (carbohydrates, proteins and fats) and micronutrients (vitamins and minerals) affecting intestinal homeostasis directly or indirectly (polysaccharides-intestinal bacteria, proteins-amino acids). In addition, new perspectives of IOs in combination with advanced biological techniques and their applications in precise nutrition were proposed.
Collapse
Affiliation(s)
- Duoduo Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China.
| | - Wengan Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China
| | - Steve W Cui
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China; Agriculture and Agri-Food Canada, Guelph Research and Development Centre, 93 Stone Road West, Guelph, Ontario NIG 5C9, Canada
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
28
|
Zhao Y, Zou DW. Gut microbiota and irritable bowel syndrome. J Dig Dis 2023; 24:312-320. [PMID: 37458142 DOI: 10.1111/1751-2980.13204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/11/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that poses a significant health concern. Although its etiology remains unknown, there is growing evidence that gut dysbiosis is involved in the development and exacerbation of IBS. Previous studies have reported altered microbial diversity, abundance, and composition in IBS patients when compared to controls. However, whether dysbiosis or aberrant changes in the intestinal microbiota can be used as a hallmark of IBS remains inconclusive. We reviewed the literatures on changes in and roles of intestinal microbiota in relation to IBS and discussed various gut microbiota manipulation strategies. Gut microbiota may affect IBS development by regulating the mucosal immune system, brain-gut-microbiome interaction, and intestinal barrier function. The advent of high-throughput multi-omics provides important insights into the pathogenesis of IBS and promotes the development of individualized treatment for IBS. Despite advances in currently available microbiota-directed therapies, large-scale, well-organized, and long-term randomized controlled trials are highly warranted to assess their clinical effects. Overall, gut microbiota alterations play a critical role in the pathophysiology of IBS, and modulation of microbiota has a significant therapeutic potential that requires to be further verified.
Collapse
Affiliation(s)
- Ye Zhao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Duo Wu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Li X, Zheng P, Cao W, Cao Y, She X, Yang H, Ma K, Wu F, Gao X, Fu Y, Yin J, Wei F, Jiang S, Cui B. Lactobacillus rhamnosus GG ameliorates noise-induced cognitive deficits and systemic inflammation in rats by modulating the gut-brain axis. Front Cell Infect Microbiol 2023; 13:1067367. [PMID: 37180445 PMCID: PMC10169735 DOI: 10.3389/fcimb.2023.1067367] [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: 10/11/2022] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
Background Environmental noise exposure is linked to neuroinflammation and imbalance of the gut microbiota. Promoting gut microbiota homeostasis may be a key factor in relieving the deleterious non-auditory effects of noise. This study aimed to investigate the effect of Lactobacillus rhamnosus GG (LGG) intervention on noise-induced cognitive deficits and systemic inflammation in rats. Methods Learning and memory were assessed using the Morris water maze, while 16S rRNA sequencing and gas chromatography-mass spectrometry were used to analyze the gut microbiota and short-chain fatty acid (SCFA) content. Endothelial tight junction proteins and serum inflammatory mediators were assessed to explore the underlying pathological mechanisms. Results The results indicated that Lactobacillus rhamnosus GG intervention ameliorated noise-induced memory deterioration, promoted the proliferation of beneficial bacteria, inhibited the growth of harmful bacteria, improved dysregulation of SCFA-producing bacteria, and regulated SCFA levels. Mechanistically, noise exposure led to a decrease in tight junction proteins in the gut and hippocampus and an increase in serum inflammatory mediators, which were significantly alleviated by Lactobacillus rhamnosus GG intervention. Conclusion Taken together, Lactobacillus rhamnosus GG intervention reduced gut bacterial translocation, restored gut and blood-brain barrier functions, and improved gut bacterial balance in rats exposed to chronic noise, thereby protecting against cognitive deficits and systemic inflammation by modulating the gut-brain axis.
Collapse
Affiliation(s)
- Xiaofang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health and Management, Binzhou Medical University, Yantai, China
| | - Pengfang Zheng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Wa Cao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Yang Cao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kefeng Ma
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Fangshan Wu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yu Fu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Jiayi Yin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University, Jinan, China
| | - Fei Wei
- School of Public Health and Management, Binzhou Medical University, Yantai, China
| | - Shoufang Jiang
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School of Public Health and Management, Binzhou Medical University, Yantai, China
- School of Public Health and Management, Weifang Medical University, Weifang, China
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University, Jinan, China
| |
Collapse
|
30
|
Lechuga S, Braga-Neto MB, Naydenov NG, Rieder F, Ivanov AI. Understanding disruption of the gut barrier during inflammation: Should we abandon traditional epithelial cell lines and switch to intestinal organoids? Front Immunol 2023; 14:1108289. [PMID: 36875103 PMCID: PMC9983034 DOI: 10.3389/fimmu.2023.1108289] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Disruption of the intestinal epithelial barrier is a hallmark of mucosal inflammation. It increases exposure of the immune system to luminal microbes, triggering a perpetuating inflammatory response. For several decades, the inflammatory stimuli-induced breakdown of the human gut barrier was studied in vitro by using colon cancer derived epithelial cell lines. While providing a wealth of important data, these cell lines do not completely mimic the morphology and function of normal human intestinal epithelial cells (IEC) due to cancer-related chromosomal abnormalities and oncogenic mutations. The development of human intestinal organoids provided a physiologically-relevant experimental platform to study homeostatic regulation and disease-dependent dysfunctions of the intestinal epithelial barrier. There is need to align and integrate the emerging data obtained with intestinal organoids and classical studies that utilized colon cancer cell lines. This review discusses the utilization of human intestinal organoids to dissect the roles and mechanisms of gut barrier disruption during mucosal inflammation. We summarize available data generated with two major types of organoids derived from either intestinal crypts or induced pluripotent stem cells and compare them to the results of earlier studies with conventional cell lines. We identify research areas where the complementary use of colon cancer-derived cell lines and organoids advance our understanding of epithelial barrier dysfunctions in the inflamed gut and identify unique questions that could be addressed only by using the intestinal organoid platforms.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Manuel B. Braga-Neto
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
31
|
Lactobacillus rhamnosus GG Promotes Recovery of the Colon Barrier in Septic Mice through Accelerating ISCs Regeneration. Nutrients 2023; 15:nu15030672. [PMID: 36771378 PMCID: PMC9921111 DOI: 10.3390/nu15030672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
Disruption of the intestinal barrier is both the cause and result of sepsis. The proliferation and differentiation of intestinal stem cells (ISCs) promote the regenerative nature of intestinal epithelial cells, repairing the injured intestinal mucosal barrier; however, it is uncertain whether the recovery effects mediated by the ISCs are related to the gut microbiota. This research found that the survival rate of septic mice was improved with a Lactobacillus rhamnosus GG (LGG) treatment. Furthermore, an increased proliferation and decreased apoptosis in colon epithelial cells were observed in the LGG-treated septic mice. In vitro, we found that a LGG supernatant was effective in maintaining the colonoid morphology and proliferation under the damage of TNF-α. Both in the mice colon and the colonoid, the LGG-induced barrier repair process was accompanied by an increased expression of Lgr5+ and lysozyme+ cells. This may be attributed to the upregulation of the IL-17, retinol metabolism, NF-kappa B and the MAPK signaling pathways, among which, Tnfaip3 and Nfkbia could be used as two potential biomarkers for LGG in intestinal inflammation therapy. In conclusion, our finding suggests that LGG protects a sepsis-injured intestinal barrier by promoting ISCs regeneration, highlighting the protective mechanism of oral probiotic consumption in sepsis.
Collapse
|
32
|
Ahn JS, Kang MJ, Seo Y, Kim HS. Intestinal organoids as advanced modeling platforms to study the role of host-microbiome interaction in homeostasis and disease. BMB Rep 2023; 56:15-23. [PMID: 36379514 PMCID: PMC9887104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/28/2023] Open
Abstract
After birth, animals are colonized by a diverse community of microorganisms. The digestive tract is known to contain the largest number of microbiome in the body. With emergence of the gut-brain axis, the importance of gut microbiome and its metabolites in host health has been extensively studied in recent years. The establishment of organoid culture systems has contributed to studying intestinal pathophysiology by replacing current limited models. Owing to their architectural and functional complexity similar to a real organ, co-culture of intestinal organoids with gut microbiome can provide mechanistic insights into the detrimental role of pathobiont and the homeostatic function of commensal symbiont. Here organoid-based bacterial co-culture techniques for modeling host-microbe interactions are reviewed. This review also summarizes representative studies that explore impact of enteric microorganisms on intestinal organoids to provide a better understanding of host-microbe interaction in the context of homeostasis and disease. [BMB Reports 2023; 56(1): 15-23].
Collapse
Affiliation(s)
- Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
33
|
Ballegaard ASR, Bøgh KL. Intestinal protein uptake and IgE-mediated food allergy. Food Res Int 2023; 163:112150. [PMID: 36596102 DOI: 10.1016/j.foodres.2022.112150] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Food allergy is affecting 5-8% of young children and 2-4% of adults and seems to be increasing in prevalence. The cause of the increase in food allergy is largely unknown but proposed to be influenced by both environmental and lifestyle factors. Changes in intestinal barrier functions and increased uptake of dietary proteins have been suggested to have a great impact on food allergy. In this review, we aim to give an overview of the gastrointestinal digestion and intestinal barrier function and provide a more detailed description of intestinal protein uptake, including the various routes of epithelial transport, how it may be affected by both intrinsic and extrinsic factors, and the relation to food allergy. Further, we give an overview of in vitro, ex vivo and in vivo techniques available for evaluation of intestinal protein uptake and gut permeability in general. Proteins are digested by gastric, pancreatic and integral brush border enzymes in order to allow for sufficient nutritional uptake. Absorption and transport of dietary proteins across the epithelial layer is known to be dependent on the physicochemical properties of the proteins and their digestion fragments themselves, such as size, solubility and aggregation status. It is believed, that the greater an amount of intact protein or larger peptide fragments that is transported through the epithelial layer, and thus encountered by the mucosal immune system in the gut, the greater is the risk of inducing an adverse allergic response. Proteins may be absorbed across the epithelial barrier by means of various mechanisms, and studies have shown that a transcellular facilitated transport route unique for food allergic individuals are at play for transport of allergens, and that upon mediator release from mast cells an enhanced allergen transport via the paracellular route occurs. This is in contrast to healthy individuals where transcytosis through the enterocytes is the main route of protein uptake. Thus, knowledge on factors affecting intestinal barrier functions and methods for the determination of their impact on protein uptake may be useful in future allergenicity assessments and for development of future preventive and treatment strategies.
Collapse
Affiliation(s)
| | - Katrine Lindholm Bøgh
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
34
|
Ahn JS, Kang MJ, Seo Y, Kim HS. Intestinal organoids as advanced modeling platforms to study the role of host-microbiome interaction in homeostasis and disease. BMB Rep 2023; 56:15-23. [PMID: 36379514 PMCID: PMC9887104 DOI: 10.5483/bmbrep.2022-0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/09/2025] Open
Abstract
After birth, animals are colonized by a diverse community of microorganisms. The digestive tract is known to contain the largest number of microbiome in the body. With emergence of the gut-brain axis, the importance of gut microbiome and its metabolites in host health has been extensively studied in recent years. The establishment of organoid culture systems has contributed to studying intestinal pathophysiology by replacing current limited models. Owing to their architectural and functional complexity similar to a real organ, co-culture of intestinal organoids with gut microbiome can provide mechanistic insights into the detrimental role of pathobiont and the homeostatic function of commensal symbiont. Here organoid-based bacterial co-culture techniques for modeling host-microbe interactions are reviewed. This review also summarizes representative studies that explore impact of enteric microorganisms on intestinal organoids to provide a better understanding of host-microbe interaction in the context of homeostasis and disease. [BMB Reports 2023; 56(1): 15-23].
Collapse
Affiliation(s)
- Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
35
|
Knox EG, Lynch CMK, Lee YS, O'Driscoll CM, Clarke G, Cryan JF, Aburto MR. The gut microbiota is important for the maintenance of blood-cerebrospinal fluid barrier integrity. Eur J Neurosci 2023; 57:233-241. [PMID: 36453579 PMCID: PMC10107533 DOI: 10.1111/ejn.15878] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
The gut microbiota communicates with the brain through several pathways including the vagus nerve, immune system, microbial metabolites and through the endocrine system. Pathways along the humoral/immune gut microbiota-brain axis are composed of a series of vascular and epithelial barriers including the intestinal epithelial barrier, gut-vascular barrier, blood-brain barrier and blood-cerebrospinal fluid barrier. Of these barriers, the relationship between the gut microbiota and blood-cerebrospinal fluid barrier is yet to be fully defined. Here, using a germ-free mouse model, we aimed to assess the relationship between the gut microbiota and the integrity of the blood-cerebrospinal fluid barrier, which is localized to the choroid plexus epithelium. Using confocal microscopy, we visualized the tight junction protein zonula occludens-1, an integral aspect of choroid plexus integrity, as well as the choroid plexus fenestrated capillaries. Quantification of tight junction proteins via network analysis led to the observation that there was a decrease in the zonula occludens-1 network organization in germ-free mice; however, we did not observe any differences in capillary structure. Taken together, these data indicate that the blood-cerebrospinal fluid barrier is another barrier along the gut microbiota-brain axis. Future studies are required to elucidate its relative contribution in signalling from microbiota to the brain.
Collapse
Affiliation(s)
- Emily G Knox
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Caoimhe M K Lynch
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Ye Seul Lee
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Maria R Aburto
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Deng Z, Dai J, Wei Y, Ma Y, Mao Y, Zhang J, Hua W, Wang H. Comparison between Lactobacillus rhamnosus GG and LuxS-deficient strain in regulating gut barrier function and inflammation in early-weaned piglets. Front Immunol 2022; 13:1080789. [PMID: 36569920 PMCID: PMC9773554 DOI: 10.3389/fimmu.2022.1080789] [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: 10/26/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
Background Early weaning-induced stress impairs the intestinal barrier function and adversely affects the health of piglet. Probiotics can be used to prevent and treat various intestinal diseases. Lactobacillus rhamnosus GG (LGG) has an LuxS/AI-2 quorum sensing (QS) system that senses environmental changes through chemical signaling molecules. The aim of the study was to explore whether luxS mutant affects the protective role of LGG in the gut barrier of weaned piglets by comparing the luxS mutant (ΔluxS) with its wild-type (WT). Methods Newborn piglets were orally administered with WT and ΔluxS at dosage of 109 CFU, respectively. Accordingly, newborn piglets in the Con group were orally administered with PBS. Piglets were weaned on day 21 and euthanized on day 24, three days following weaning. Results Supplementation of ΔluxS in advance significantly boosted the relative abundances of healthy microbes (including Catenibacterium, Eubacterium, Lachnospiraceae and Bifidobacterium). WT and ΔluxS maintain intestinal barrier function mainly by promoting intestinal villus to crypt ratio (VCR), occludin protein expression and mucus secretion (P<0.05). Furthermore, LGG reduces pro-inflammatory mediators by inhibiting TLR4 and MAPK signal transduction (P<0.05). Conclusion Both WT and ΔluxS were shown to resist weaning stress by enhancing the intestinal barrier function of piglets. It has to be said that the ability of ΔluxS to maintain intestinal tissue morphology and promote mucus secretion significantly decreased compared with that of WT.
Collapse
Affiliation(s)
- Zhaoxi Deng
- Experimental Livestock Farm of Animal Husbandry and Veterinary Research Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China,College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China,Laboratory Animal Center, Sichuan University, Chengdu, China
| | - Jinyan Dai
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Yusen Wei
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Yanfei Ma
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Yingying Mao
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Jinzhi Zhang
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | - Weidong Hua
- Experimental Livestock Farm of Animal Husbandry and Veterinary Research Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China,*Correspondence: Weidong Hua, ; Haifeng Wang,
| | - Haifeng Wang
- College of Animal Science, Ministry of Education (MOE) Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China,*Correspondence: Weidong Hua, ; Haifeng Wang,
| |
Collapse
|
37
|
Luo M, Liu Q, Xiao L, Xiong LS. Golden bifid might improve diarrhea-predominant irritable bowel syndrome via microbiota modulation. JOURNAL OF HEALTH, POPULATION AND NUTRITION 2022; 41:21. [PMID: 35578355 PMCID: PMC9109320 DOI: 10.1186/s41043-022-00302-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Objective Gut microbiota might play a crucial role in the pathogenesis of irritable bowel syndrome (IBS), and probiotics supplement may be an effective treatment option. This study aims to explore the therapeutic effects of Golden bifid on the diarrhea-predominant IBS (IBS-D). Methods Twenty-one consecutive IBS-D patients were recruited based on Rome IV criteria. All patients took 2000 mg Golden bifid triple daily for 4 weeks. Gastrointestinal (GI) symptoms, psychological symptoms, small intestine bacterial overgrowth (SIBO) and fecal microbiota characteristics were evaluated in IBS-D patients before and after treatment. Results After 4-week treatment of Golden bifid, the GI symptoms such as abdominal pain (2.90 ± 1.04 vs. 1.90 ± 1.26, P = 0.002), abdominal distension (2.00 ± 1.34 vs. 1.29 ± 1.31, P = 0.007), diarrhea (3.24 ± 1.37 vs. 1.81 ± 1.21, P = 0.001), defecatory urgency (3.48 ± 1.03 vs. 2.33 ± 1.35, P = 0.000) and incomplete evacuation (2.71 ± 1.15 vs. 1.76 ± 1.26, P = 0.003) were significantly alleviated in IBS-D patients. The Self-Rating Depression Scale (SDS) decreased significantly (46.19 ± 11.36 vs. 43.33 ± 9.65, P = 0.041), and SIBO could be eradicated in 25% (4/16) of IBS-D patients with SIBO. Meanwhile, the abundance of Unclassified Lachnospiraceae and Dorea in genus level and Unclassified Lachnospiraceae, Bacterium Dorea, Bacterium Butyricicoccus and Dorea formicigenerans ATCC 27755 in species level were increased in fecal microbiota (P < 0.05). Conclusions Golden bifid could improve most of GI symptoms and depressive symptoms in IBS-D patients and eradicate a small proportion of SIBO in those IBS-D patients with SIBO. What's more, Golden bifid could also modulate the fecal microbiota in IBS-D patients, which implied that the Golden bifid might improve IBS-D via microbiota modulation. Supplementary Information The online version contains supplementary material available at 10.1186/s41043-022-00302-0.
Collapse
|
38
|
Mavrogeni ME, Asadpoor M, Henricks PAJ, Keshavarzian A, Folkerts G, Braber S. Direct Action of Non-Digestible Oligosaccharides against a Leaky Gut. Nutrients 2022; 14:4699. [PMID: 36364961 PMCID: PMC9655944 DOI: 10.3390/nu14214699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 10/28/2023] Open
Abstract
The epithelial monolayer is the primary determinant of mucosal barrier function, and tight junction (TJ) complexes seal the paracellular space between the adjacent epithelial cells and represent the main "gate-keepers" of the paracellular route. Impaired TJ functionality results in increased permeation of the "pro-inflammatory" luminal contents to the circulation that induces local and systemic inflammatory and immune responses, ultimately triggering and/or perpetuating (chronic) systemic inflammatory disorders. Increased gut leakiness is associated with intestinal and systemic disease states such as inflammatory bowel disease and neurodegenerative diseases such as Parkinson's disease. Modulation of TJ dynamics is an appealing strategy aiming at inflammatory conditions associated with compromised intestinal epithelial function. Recently there has been a growing interest in nutraceuticals, particularly in non-digestible oligosaccharides (NDOs). NDOs confer innumerable health benefits via microbiome-shaping and gut microbiota-related immune responses, including enhancement of epithelial barrier integrity. Emerging evidence supports that NDOs also exert health-beneficial effects on microbiota independently via direct interactions with intestinal epithelial and immune cells. Among these valuable features, NDOs promote barrier function by directly regulating TJs via AMPK-, PKC-, MAPK-, and TLR-associated pathways. This review provides a comprehensive overview of the epithelial barrier-protective effects of different NDOs with a special focus on their microbiota-independent modulation of TJs.
Collapse
Affiliation(s)
- Maria Eleni Mavrogeni
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Ali Keshavarzian
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
39
|
Role of Intestinal Microbes in Chronic Liver Diseases. Int J Mol Sci 2022; 23:ijms232012661. [PMID: 36293518 PMCID: PMC9603943 DOI: 10.3390/ijms232012661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022] Open
Abstract
With the recent availability and upgrading of many emerging intestinal microbes sequencing technologies, our research on intestinal microbes is changing rapidly. A variety of investigations have found that intestinal microbes are essential for immune system regulation and energy metabolism homeostasis, which impacts many critical organs. The liver is the first organ to be traversed by the intestinal portal vein, and there is a strong bidirectional link between the liver and intestine. Many intestinal factors, such as intestinal microbes, bacterial composition, and intestinal bacterial metabolites, are deeply involved in liver homeostasis. Intestinal microbial dysbiosis and increased intestinal permeability are associated with the pathogenesis of many chronic liver diseases, such as alcoholic fatty liver disease (AFLD), non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), chronic hepatitis B (CHB), chronic hepatitis C (CHC), autoimmune liver disease (AIH) and the development of hepatocellular carcinoma (HCC). Intestinal permeability and dysbacteriosis often lead to Lipopolysaccharide (LPS) and metabolites entering in serum. Then, Toll-like receptors activation in the liver induces the exposure of the intestine and liver to many small molecules with pro-inflammatory properties. And all of these eventually result in various liver diseases. In this paper, we have discussed the current evidence on the role of various intestinal microbes in different chronic liver diseases. As well as potential new therapeutic approaches are proposed in this review, such as antibiotics, probiotics, and prebiotics, which may have an improvement in liver diseases.
Collapse
|
40
|
Iribarren C, Nordlander S, Sundin J, Isaksson S, Savolainen O, Törnblom H, Magnusson MK, Simrén M, Öhman L. Fecal luminal factors from patients with irritable bowel syndrome induce distinct gene expression of colonoids. Neurogastroenterol Motil 2022; 34:e14390. [PMID: 35485994 PMCID: PMC9786662 DOI: 10.1111/nmo.14390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alteration of the host-microbiota cross talk at the intestinal barrier may participate in the pathophysiology of irritable bowel syndrome (IBS). Therefore, we aimed to determine effects of fecal luminal factors from IBS patients on the colonic epithelium using colonoids. METHODS Colon-derived organoid monolayers, colonoids, generated from a healthy subject, underwent stimulation with fecal supernatants from healthy subjects and IBS patients with predominant diarrhea, phosphate-buffered saline (PBS), or lipopolysaccharide (LPS). Cytokines in cell cultures and fecal LPS were measured by ELISA and mRNA gene expression of monolayers was analyzed using Qiagen RT2 Profiler PCR Arrays. The fecal microbiota profile was determined by the GA-map™ dysbiosis test and the fecal metabolite profile was analyzed by untargeted liquid chromatography/mass spectrometry. KEY RESULTS Colonoid monolayers stimulated with fecal supernatants from healthy subjects (n = 7), PBS (n = 4) or LPS (n = 3) presented distinct gene expression profiles, with some overlap (R2 Y = 0.70, Q2 = 0.43). Addition of fecal supernatants from healthy subjects and IBS patients (n = 9) gave rise to different gene expression profiles of the colonoid monolayers (R2 Y = 0.79, Q2 = 0.64). Genes (n = 22) related to immune response (CD1D, TLR5) and barrier integrity (CLDN15, DSC2) contributed to the separation. Levels of proinflammatory cytokines in colonoid monolayer cultures were comparable when stimulated with fecal supernatants from either donor types. Fecal microbiota and metabolite profiles, but not LPS content, differed between the study groups. CONCLUSIONS Fecal luminal factors from IBS patients induce a distinct colonic epithelial gene expression, potentially reflecting the disease pathophysiology. The culture of colonoids from healthy subjects with fecal supernatants from IBS patients may facilitate the exploration of IBS related intestinal micro-environmental and barrier interactions.
Collapse
Affiliation(s)
- Cristina Iribarren
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden,Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Sofia Nordlander
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden,Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Johanna Sundin
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Stefan Isaksson
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Otto Savolainen
- Chalmers Mass Spectrometry InfrastructureDepartment of Biology and Biological EngineeringChalmers University of TechnologyGothenburgSweden
| | - Hans Törnblom
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria K. Magnusson
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Magnus Simrén
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden,Center for Functional GI and Motility DisordersUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Lena Öhman
- Department of Microbiology and ImmunologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
41
|
Li K, Cheng X, Jin R, Han T, Li J. The influence of different proton pump inhibitors and potassium-competitive acid blockers on indomethacin-induced small intestinal injury. J Gastroenterol Hepatol 2022; 37:1935-1945. [PMID: 35938741 DOI: 10.1111/jgh.15973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/18/2022] [Accepted: 08/03/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM The influence of gastric acid inhibitors (GAIs) on nonsteroidal anti-inflammatory drug (NSAID)-induced enteropathy is controversial. Herein, the influences of different GAIs on NSAID-induced intestinal injury and the underlying mechanisms are clarified. METHODS Indomethacin (IND; 10 mg/kg/day) was administered to mice to induce small intestinal injury. Disease activity was examined macroscopically and histologically. The permeability of small intestine was evaluated by measuring plasma lipopolysaccharide levels. 16S rDNA sequencing was performed to determine the composition of intestinal flora. RESULTS Among the four GAIs, ilaprazole (IPZ) significantly attenuated IND-induced small intestinal injury and maintained the integrity of the mucosal barrier. Omeprazole (OPZ) and vonoprazan (VPZ) ameliorated ulceration without significant differences, while rabeprazole (RPZ) failed to protect against the injury. To explore the potential mechanism, we investigated changes in the gut microbiota mediated by GAIs. After 5-day administration, GAIs significantly altered the composition of the gut microbiota. The IND group had a significant decrease in alpha diversity compared with the control group, and this decrease was reversed by OPZ and IPZ treatment, respectively. After IPZ treatment, the community membership was more assembled in the control group than the IND group. Further, we found that Lactobacillus was significantly increased in the groups of OPZ, IPZ, and VPZ, while Bacteroides was significantly increased in the RPZ group. CONCLUSION Our results indicated that GAIs have different influences on the mucosal barrier, possibly by altering the composition of intestinal microbiota, and the impacts mediated by various GAIs in the IND-induced intestinal damage model seem different.
Collapse
Affiliation(s)
- Kemin Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyun Cheng
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Jin
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Taotao Han
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Zheng J, Ahmad AA, Yang Y, Liang Z, Shen W, Feng M, Shen J, Lan X, Ding X. Lactobacillus rhamnosus CY12 Enhances Intestinal Barrier Function by Regulating Tight Junction Protein Expression, Oxidative Stress, and Inflammation Response in Lipopolysaccharide-Induced Caco-2 Cells. Int J Mol Sci 2022; 23:ijms231911162. [PMID: 36232464 PMCID: PMC9569798 DOI: 10.3390/ijms231911162] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
The intestinal barrier is vital for preventing inflammatory bowel disease (IBD). The objectives of this study were to assess whether the Lactobacillus rhamnosus CY12 could alleviate oxidative stress, inflammation, and the disruption of tight junction (TJ) barrier functions induced by lipopolysaccharide (LPS), and therefore to explore the potential underlying molecular mechanisms. Our results showed that LPS-induced Cancer coli-2 (Caco-2) cells significantly increased the levels of reactive oxygen species (ROS), lactate dehydrogenase, inflammatory cytokines interleukin-1β, interleukin-6, interleukin-8, and tumor necrosis factor-α (IL-1β, IL-6, IL-8, and TNF-α), and the cell apoptosis rate while decreasing the levels of TJ proteins occludin, zonula occludens-1 (ZO-1), and claudin and antioxidant enzymes, such as catalase, superoxide dismutase, and glutathione peroxidase(CAT, SOD, and GSH-Px) (p < 0.05). However, Lactobacillus rhamnosus CY12 could relieve cytotoxicity, apoptosis, oxidative stress, and pro-inflammatory cytokine expressions, and also inhibit the Toll-like receptor 4/nuclear factor kappa-B(TLR4/NF-κB) signaling pathway. Furthermore, the gene expression of antioxidant enzymes, as well as the mRNA and protein expressions of TJ proteins, was improved. Particularly, the concentration of 108 cfu/mL significantly prevented the inflammatory injury induced by LPS in Caco-2 cells (p < 0.05). These findings support a potential application of Lactobacillus rhamnosus CY12 as a probiotic to prevent LPS-induced intestinal injury and treat intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Juanshan Zheng
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Anum Ali Ahmad
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- State Key Laboratory of Grassland Agro-Ecosystems, School of Life Sciences, Lanzhou University, Lanzhou 730050, China
| | - Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Zeyi Liang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Min Feng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Jiahao Shen
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
| | - Xianyong Lan
- Laboratory of Animal Genome and Gene Function, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| | - Xuezhi Ding
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Academy of Agricultural Sciences, Lanzhou 730050, China
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China
- Correspondence: (X.L.); (X.D.); Tel.: +86-931-211-5255 (X.D.)
| |
Collapse
|
43
|
Günther C, Winner B, Neurath MF, Stappenbeck TS. Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut 2022; 71:1892-1908. [PMID: 35636923 PMCID: PMC9380493 DOI: 10.1136/gutjnl-2021-326560] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
We are entering an era of medicine where increasingly sophisticated data will be obtained from patients to determine proper diagnosis, predict outcomes and direct therapies. We predict that the most valuable data will be produced by systems that are highly dynamic in both time and space. Three-dimensional (3D) organoids are poised to be such a highly valuable system for a variety of gastrointestinal (GI) diseases. In the lab, organoids have emerged as powerful systems to model molecular and cellular processes orchestrating natural and pathophysiological human tissue formation in remarkable detail. Preclinical studies have impressively demonstrated that these organs-in-a-dish can be used to model immunological, neoplastic, metabolic or infectious GI disorders by taking advantage of patient-derived material. Technological breakthroughs now allow to study cellular communication and molecular mechanisms of interorgan cross-talk in health and disease including communication along for example, the gut-brain axis or gut-liver axis. Despite considerable success in culturing classical 3D organoids from various parts of the GI tract, some challenges remain to develop these systems to best help patients. Novel platforms such as organ-on-a-chip, engineered biomimetic systems including engineered organoids, micromanufacturing, bioprinting and enhanced rigour and reproducibility will open improved avenues for tissue engineering, as well as regenerative and personalised medicine. This review will highlight some of the established methods and also some exciting novel perspectives on organoids in the fields of gastroenterology. At present, this field is poised to move forward and impact many currently intractable GI diseases in the form of novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Stem Cell Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
44
|
Bai Y, Ma K, Li J, Ren Z, Zhang J, Shan A. Lactobacillus rhamnosus GG ameliorates DON-induced intestinal damage depending on the enrichment of beneficial bacteria in weaned piglets. J Anim Sci Biotechnol 2022; 13:90. [PMID: 35962456 PMCID: PMC9375241 DOI: 10.1186/s40104-022-00737-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deoxynivalenol (DON) is one of the most common environmental pollutants that induces intestinal inflammation and microbiota dysbiosis. Lactobacillus rhamnosus GG (LGG) is a probiotic that not only has anti-inflammatory effects, but also shows protective effect on the intestinal barrier. However, it is still unknown whether LGG exerts beneficial effects against DON-induced intestinal damage in piglets. In this work, a total of 36 weaned piglets were randomized to one of four treatment groups for 21 d. The treatment groups were CON (basal diet); LGG (basal diet supplemented with 1.77 × 1011 CFU/kg LGG); DON (DON-contaminated diet) and LGG + DON (DON-contaminated diet supplemented with 1.77 × 1011 CFU/kg LGG). RESULT Supplementation of LGG can enhance growth performance of piglets exposed to DON by improving intestinal barrier function. LGG has a mitigating effect on intestinal inflammation induced by DON exposure, largely through repression of the TLR4/NF-κB signaling pathway. Furthermore, supplementation of LGG increased the relative abundances of beneficial bacteria (e.g., Collinsella, Lactobacillus, Ruminococcus_torques_group and Anaerofustis), and decreased the relative abundances of harmful bacteria (e.g., Parabacteroides and Ruminiclostridium_6), and also promoted the production of SCFAs. CONCLUSIONS LGG ameliorates DON-induced intestinal damage, which may provide theoretical support for the application of LGG to alleviate the adverse effects induced by DON exposure.
Collapse
Affiliation(s)
- Yongsong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Kaidi Ma
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Zhongshuai Ren
- College of Animal Sciences, Jilin University, Key Laboratory of Zoonosis Research, Ministry of Education, Changchun, 130062, P. R. China
| | - Jing Zhang
- College of Animal Sciences, Jilin University, Key Laboratory of Zoonosis Research, Ministry of Education, Changchun, 130062, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| |
Collapse
|
45
|
Chen G, Shi F, Yin W, Guo Y, Liu A, Shuai J, Sun J. Gut microbiota dysbiosis: The potential mechanisms by which alcohol disrupts gut and brain functions. Front Microbiol 2022; 13:916765. [PMID: 35966709 PMCID: PMC9372561 DOI: 10.3389/fmicb.2022.916765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/29/2022] [Indexed: 11/24/2022] Open
Abstract
Alcohol use disorder (AUD) is a high-risk psychiatric disorder and a key cause of death and disability in individuals. In the development of AUD, there is a connection known as the microbiota-gut-brain axis, where alcohol use disrupts the gut barrier, resulting in changes in intestinal permeability as well as the gut microbiota composition, which in turn impairs brain function and worsens the patient’s mental status and gut activity. Potential mechanisms are explored by which alcohol alters gut and brain function through the effects of the gut microbiota and their metabolites on immune and inflammatory pathways. Alcohol and microbiota dysregulation regulating neurotransmitter release, including DA, 5-HT, and GABA, are also discussed. Thus, based on the above discussion, it is possible to speculate on the gut microbiota as an underlying target for the treatment of diseases associated with alcohol addiction. This review will focus more on how alcohol and gut microbiota affect the structure and function of the gut and brain, specific changes in the composition of the gut microbiota, and some measures to mitigate the changes caused by alcohol exposure. This leads to a potential intervention for alcohol addiction through fecal microbiota transplantation, which could normalize the disruption of gut microbiota after AUD.
Collapse
Affiliation(s)
- Ganggang Chen
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Fenglei Shi
- Department of Othopaedics, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Yin
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Yao Guo
- Shandong Provincial Mental Health Center, Jinan, China
| | - Anru Liu
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jiacheng Shuai
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
| | - Jinhao Sun
- Department of Anatomy and Neurobiology, School of Basic Medicine, Shandong University, Jinan, China
- *Correspondence: Jinhao Sun,
| |
Collapse
|
46
|
Kim MB, Hwangbo S, Jang S, Jo YK. Bioengineered Co-culture of organoids to recapitulate host-microbe interactions. Mater Today Bio 2022; 16:100345. [PMID: 35847376 PMCID: PMC9283667 DOI: 10.1016/j.mtbio.2022.100345] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/05/2022] Open
Abstract
The recent spike in the instances of complex physiological host-microbe interactions has raised the demand for developing in vitro models that recapitulate the microbial microenvironment in the human body. Organoids are steadily emerging as an in vitro culture system that closely mimics the structural, functional, and genetic features of complex human organs, particularly for better understanding host-microbe interactions. Recent advances in organoid culture technology have become new avenues for assessing the pathogenesis of symbiotic interactions, pathogen-induced infectious diseases, and various other diseases. The co-cultures of organoids with microbes have shown great promise in simulating host-microbe interactions with a high level of complexity for further advancement in related fields. In this review, we provide an overview of bioengineering approaches for microbe-co-cultured organoids. Latest developments in the applications of microbe-co-cultured organoids to study human physiology and pathophysiology are also highlighted. Further, an outlook on future research on bioengineered organoid co-cultures for various applications is presented.
Collapse
|
47
|
Effects of different probiotic strains B. lactis, L. rhamnosus and L. reuteri on brain-intestinal axis immunomodulation in an endotoxin-induced inflammation. Mol Neurobiol 2022; 59:5168-5178. [PMID: 35674863 DOI: 10.1007/s12035-022-02906-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
Abstract
The study evaluated the effects of supplementation with three different probiotic strains Bifidobacterium lactis (LACT GB™), Lactobacillus rhamnosus (RHAM GB™) and Lactobacillus reuteri (REUT GB™) on brain-intestinal immunomodulation in an animal model of LPS-induced inflammation. Fifty mice Balb/C were distributed into five groups: control; lipopolysaccharide (LPS); LPS + B. lactis (LACT GB™); LPS + L. rhamnosus (RHAM GB™); and LPS + L. reuteri (REUT GB™). The animals were supplemented with their respective probiotic microorganisms daily, for 30 days, at a concentration of 1 × 109 CFU/animal/day. After 30 days of supplementation, animals received the inflammatory insult by LPS (15 mg/kg). Behavioral tests, oxidative stress and inflammation were performed, as well as gut and brain histology. In the behavioral test, LPS + B. lactis group was less anxious than the other groups. Serum interleukin IL-1β and IL-6 levels increased in all groups that received the LPS insult, and there was a reduction in inflammation in the supplemented groups when compared to the LPS group in brain and gut. There is a reduction in myeloperoxidase activity and oxidative stress in groups supplemented with probiotics. In intestine histological analysis occurs damage to the tissue integrity in the LPS group, in the other hand, occurs preservation of integrity in the probiotic supplemented animals. In the brain, infiltrates of perivascular inflammatory cells can be seen in the LPS group. The three probiotic studies showed efficient immunomodulating activity and ensured integrity of the intestinal barrier function, even after the severe insult by LPS. These results show the important role of probiotics in the gut-brain axis. Graphical abstract illustratively represents the gut-brain axis and how different probiotic strains influence the immunomodulatory response releasing different pro- and anti-inflammatory cytokines, and their role in the balance of dysbiosis.
Collapse
|
48
|
Pharmacological Treatments Available for Immune-Checkpoint-Inhibitor-Induced Colitis. Biomedicines 2022; 10:biomedicines10061334. [PMID: 35740355 PMCID: PMC9219666 DOI: 10.3390/biomedicines10061334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitor treatment has shown revolutionary therapeutic effects in various carcinomas. However, immune-related adverse events (irAE) following this treatment can sometimes lead to treatment discontinuation. One such frequently encountered adverse event is immune-related colitis (irAE colitis). Corticosteroids (CS) are the first-line treatment for irAE colitis, but we often encounter CS-refractory or -resistant cases. The application of multiple biologics has been proposed as a therapy to be administered after CS treatment; however, the efficacy and safety of biologics for patients with irAE colitis who do not respond to CS have not been established. This review summarizes the treatment regimens available for irAE colitis, focusing on the mechanism of action of corticosteroids, infliximab, vedolizumab, and other drugs.
Collapse
|
49
|
Liu X, Jin G, Tang Q, Huang S, Zhang Y, Sun Y, Liu T, Guo Z, Yang C, Wang B, Jiang K, Zhong W, Cao H. Early life Lactobacillus rhamnosus GG colonisation inhibits intestinal tumour formation. Br J Cancer 2022; 126:1421-1431. [PMID: 35091695 PMCID: PMC9090826 DOI: 10.1038/s41416-021-01562-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 08/04/2021] [Accepted: 09/17/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Gut microbiota dysbiosis is closely related to the progression of colorectal cancer. Our previous study revealed that early life colonisation with Lactobacillus rhamnosus GG (LGG) had long-term positive effects on health. We sought to investigate whether early life LGG colonisation could inhibit intestinal tumour formation in offspring. METHODS Adult C57BL/6 female mice were mated with Apcmin/+ male mice. Pregnant mice with the same conception date received 108 cfu live or fixed LGG from day 18 of pregnancy until natural delivery. After genotyping, offspring mice received 107 cfu of live or fixed LGG for 0-5 days after birth. RESULTS Early life LGG colonisation significantly promoted intestinal development, inhibited low-grade intestinal inflammation and altered the gut microbiota composition of offspring in the weaning period (3 week old). Notably, early life LGG colonisation reduced the multiplicity of intestinal tumours in adulthood (12 week old), possibly due to inhibition of Wnt signalling and promotion of tumour cell apoptosis. Importantly, at the genus level, Bifidobacterium and Anaeroplasma with potential anti-tumour effects were increased in adulthood, while Peptostreptococcus, which potentially contributes to tumour formation, was decreased. CONCLUSIONS Early life LGG colonisation inhibited the intestinal tumour formation of offspring in adulthood.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Qiang Tang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Shumin Huang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yujie Zhang
- Department of Pathology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Cheng Yang
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China.
| |
Collapse
|
50
|
Singh SB, Coffman CN, Varga MG, Carroll-Portillo A, Braun CA, Lin HC. Intestinal Alkaline Phosphatase Prevents Sulfate Reducing Bacteria-Induced Increased Tight Junction Permeability by Inhibiting Snail Pathway. Front Cell Infect Microbiol 2022; 12:882498. [PMID: 35694541 PMCID: PMC9177943 DOI: 10.3389/fcimb.2022.882498] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Tight junctions (TJs) are essential components of intestinal barrier integrity and protect the epithelium against passive paracellular flux and microbial translocation. Dysfunctional TJ leads to leaky gut, a condition associated with diseases including inflammatory bowel disease (IBD). Sulfate-Reducing Bacteria (SRB) are minor residents of the gut. An increased number of Desulfovibrio, the most predominant SRB, is observed in IBD and other diseases associated with leaky gut. However, it is not known whether Desulfovibrio contributes to leaky gut. We tested the hypothesis that Desulfovibrio vulgaris (DSV) may induce intestinal permeability in vitro. Snail, a transcription factor, disrupts barrier function by affecting TJ proteins such as occludin. Intestinal alkaline phosphatase (IAP), a host defense protein, protects epithelial barrier integrity. We tested whether DSV induced permeability in polarized Caco-2 cells via snail and if this effect was inhibited by IAP. Barrier integrity was assessed by measuring transepithelial electric resistance (TEER) and by 4kDa FITC-Dextran flux to determine paracellular permeability. We found that DSV reduced TEER, increased FITC-flux, upregulated snail protein expression, caused nuclear translocation of snail, and disrupted occludin staining at the junctions. DSV-induced permeability effects were inhibited in cells knocked down for snail. Pre-treatment of cells with IAP inhibited DSV-induced FITC flux and snail expression and DSV-mediated disruption of occludin staining. These data show that DSV, a resident commensal bacterium, can contribute to leaky gut and that snail may serve as a novel therapeutic target to mitigate DSV-induced effects. Taken together, our study suggests a novel underlying mechanism of association of Desulfovibrio bloom with diseases with increased intestinal permeability. Our study also underscores IAP as a novel therapeutic intervention for correcting SRB-induced leaky gut via inhibition of snail.
Collapse
Affiliation(s)
- Sudha B. Singh
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Cristina N. Coffman
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Matthew G. Varga
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Amanda Carroll-Portillo
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Cody A. Braun
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Henry C. Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States
- Medicine Service, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
- *Correspondence: Henry C. Lin,
| |
Collapse
|