1
|
Li S, Oliva P, Zhang L, Goodrich JA, McConnell R, Conti DV, Chatzi L, Aung M. Associations between per-and polyfluoroalkyl substances (PFAS) and county-level cancer incidence between 2016 and 2021 and incident cancer burden attributable to PFAS in drinking water in the United States. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2025:10.1038/s41370-024-00742-2. [PMID: 39789195 DOI: 10.1038/s41370-024-00742-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Exposure to per- and polyfluoroalkyl substances (PFAS) has been linked with various cancers. Assessment of PFAS in drinking water and cancers can help inform biomonitoring and prevention efforts. OBJECTIVE To screen for incident cancer (2016-2021) and assess associations with PFAS contamination in drinking water in the US. METHODS We obtained county-level age-adjusted cancer incidence (2016-2021) from the Surveillance, Epidemiology, and End Results (SEER) Program. Data on PFAS levels in public drinking water systems were obtained from the Third (UCMR3; 2013-2015) and Fifth (UCMR5; 2023-2024) Unregulated Contaminant Monitoring Rule. UCMR3 measured PFOS, PFOA, PFNA, PFHxS, PFHpA, and PFBS. UCMR5 expanded measurements to include PFBA, PFHxA, PFPeA, and PFPeS. We created indicators of PFAS detection and, for UCMR5, concentrations above Maximum Contaminant Levels (MCLs). MCLs for PFOA and PFOS are 4 ng/L, and for PFNA and PFHxS are 10 ng/L. We used Poisson regression models to assess associations between PFAS detection or MCL violation and cancer incidence, adjusting for potential confounders. We estimated the number of attributable cancer cases. RESULTS PFAS in drinking water was associated with increased cancer incidence in the digestive, endocrine, oral cavity/pharynx, and respiratory systems. Incidence rate ratios (IRRs) ranged from 1.02 to 1.33. The strongest association was observed between PFBS and oral cavity/pharynx cancers (IRR: 1.33 [1.04, 1.71]). Among males, PFAS was associated with cancers in the urinary, brain, leukemia, and soft tissues. Among females, PFAS was associated with cancers in the thyroid, oral cavity/pharynx, and soft tissue. PFAS in drinking water is estimated to contribute to 4626 [95% CI: 1,377, 8046] incident cancer cases per year based on UCMR3 data and 6864 [95% CI: 991, 12,804] based on UCMR5. IMPACT STATEMENT The ecological study examined the associations between PFAS in drinking water measured in two waves (2013-2015 and 2023-2024) and cancer incidence between 2016 and 2021. We found that PFAS in drinking water was associated with cancers in the organ system including the oral cavity/pharynx, lung, digestive system, brain, urinary system, soft tissue, and thyroid. Some cancers have not been widely studied for their associations with PFAS. We also observed sex differences in the associations between PFAS and cancer risks. This is the first ecological study that examined PFAS exposure in drinking water and various cancer risks.
Collapse
Affiliation(s)
- Shiwen Li
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Paulina Oliva
- Department of Economics, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Lu Zhang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jesse A Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Max Aung
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Di Mattia M, Sallese M, Lopetuso LR. The interplay between gut microbiota and the unfolded protein response: Implications for intestinal homeostasis preservation and dysbiosis-related diseases. Microb Pathog 2025; 200:107279. [PMID: 39761770 DOI: 10.1016/j.micpath.2025.107279] [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: 03/08/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
The unfolded protein response (UPR) is a complex intracellular signal transduction system that orchestrates the cellular response during Endoplasmic Reticulum (ER) stress conditions to reestablish cellular proteostasis. If, on one side, prolonged ER stress conditions can lead to programmed cell death and autophagy as a cytoprotective mechanism, on the other, unresolved ER stress and improper UPR activation represent a perilous condition able to trigger or exacerbate inflammatory responses. Notably, intestinal and immune cells experience ER stress physiologically due to their high protein secretory rate. Indeed, there is evidence of UPR's involvement in both physiological and pathological intestinal conditions, while less is known about its bidirectional interaction with gut microbiota. However, gut microbes and their metabolites can influence ER stress and UPR pathways, and, in turn, ER stress conditions can shape gut microbiota composition, with important implications for overall intestinal health. Thus, targeting UPR components is an intriguing strategy for treating ER stress-linked dysbiosis and diseases, particularly intestinal inflammation.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
3
|
Huang X, Jiang F, Chen X, Xian Y. Plant-Derived Polysaccharides Benefit Weaned Piglets by Regulating Intestinal Microbiota: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28225-28245. [PMID: 39663725 DOI: 10.1021/acs.jafc.4c08816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
The intestine harbors a community of bacteria that is intestinal microbiota, which is a complex and highly diverse community. This review discusses the gut microbiota in piglets, including the role of intestinal homeostasis in maintaining piglet health and the various factors that influence gut microbiota. Nutritional interventions, particularly the supplementation of plant-derived polysaccharides, including dietary fiber, for weaned piglets have been shown to enhance the abundance and colonization of beneficial intestinal microbes, reduce the incidence of gastrointestinal infections, and decrease the frequency of diarrhea, thereby improving gut health and growth performance. In this context, various polysaccharides, such as those derived from Medicago sativa L. (alfalfa), Glycyrrhiza uralensis Fisch. (licorice), and Lycium barbarum L. (wolfberry), Panax ginseng C.A. Mey. (ginseng), and Astragalus membranaceus (Fisch.) Bunge (astragalus) has demonstrated significant success. Additionally, dietary fibers such as inulin, pectin, beta-glucans, gums, cellulose, resistant starch, and starch derivatives have shown potential in regulating the gastrointestinal microbiota. Research has also explored the correlation between the structural characteristics of dietary polysaccharides and their biological activities. This review will pave the way for the development and utilization of plant-derived polysaccharides as effective non-antibiotic alternatives to restore gut microbial balance in weaning piglets.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Faculty of Modern Agriculture, Yibin Vocational & Technical College, 300 Road Yuhua, District Nanxi, Yibin, Sichuan 644100, People's Republic of China
- Department of Traditional Chinese veterinary Medicine Assessment, Engineering Center of Agricultural Biosafety Assessment and Biotechnology, Yibin Vocational and Technical College, Yibin, Sichuan 644100, People's Republic of China
| | - Faming Jiang
- Faculty of Modern Agriculture, Yibin Vocational & Technical College, 300 Road Yuhua, District Nanxi, Yibin, Sichuan 644100, People's Republic of China
- Engineering Center of Agricultural Biosafety Assessment and Biotechnology, School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, Sichuan 644100, People's Republic of China
| | - Xingying Chen
- Faculty of Modern Agriculture, Yibin Vocational & Technical College, 300 Road Yuhua, District Nanxi, Yibin, Sichuan 644100, People's Republic of China
- Department of Traditional Chinese veterinary Medicine Assessment, Engineering Center of Agricultural Biosafety Assessment and Biotechnology, Yibin Vocational and Technical College, Yibin, Sichuan 644100, People's Republic of China
| | - Yuanhua Xian
- Faculty of Modern Agriculture, Yibin Vocational & Technical College, 300 Road Yuhua, District Nanxi, Yibin, Sichuan 644100, People's Republic of China
| |
Collapse
|
4
|
Marcato F, Schokker D, Kar SK, Bossers A, Harders F, Rebel JMJ, Jansen CA, van der Valk E, Kruijt L, te Beest DE, de Jong IC. Effects of breed and early feeding on intestinal microbiota, inflammation markers, and behavior of broiler chickens. Front Vet Sci 2024; 11:1492274. [PMID: 39687852 PMCID: PMC11648218 DOI: 10.3389/fvets.2024.1492274] [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: 09/06/2024] [Accepted: 10/31/2024] [Indexed: 12/18/2024] Open
Abstract
Recently, the Netherlands has shifted toward more welfare-friendly broiler production systems using slower-growing broiler breeds. Early post-hatch feeding (EF) is a dietary strategy that is currently used in commercial broiler production to modulate the gut microbiota and improve performance and welfare. However, there is a knowledge gap in how both breed and EF and their interplay affect gut microbiota composition and diversity, inflammatory status, and broiler behavior. Therefore, the aim of this study was to investigate the effects of breed (fast vs. slower-growing), EF, and their interaction on jejunum microbiota, inflammation, and behavior of broiler chickens. The study included a total of 416 Ross 308 and 416 Hubbard JA757 day-old male broiler chickens, observed until they were 37 days and 51 days old, respectively. Within each breed, one-half of the chickens received EF and the other half did not. A total of two chickens per pen were euthanized at two time points, that is, target body weight (BW) of 200 g and 2.5 kg, and jejunum samples were collected. The jejunum content samples (N = 96) were analyzed for their microbiota, whereas the jejunum tissue (N = 96) was used for the detection of mRNA levels of cytokines (IL-17, IL-22, and IFNγ). Two behavioral tests were performed to assess fear responses: (1) a novel environment test at a target BW of 200 g and (2) a tonic immobility test at a target BW of 2.5 kg. Breed affected the microbiota at a target BW of 2.5 kg (p = 0.04). A breed × EF interaction (p = 0.02) was present for IFNγ at a target BW of 200 g. During the novel environment test, Ross 308 chickens exhibited a shorter latency to vocalize and a higher number of vocalizations compared to Hubbard JA757 chickens (p < 0.05). Early-fed broiler chickens vocalized less compared to not early-fed chickens (Δ = -27.8 on average; p < 0.01). During the tonic immobility test, Hubbard JA757 chickens exhibited a shorter latency to stand compared to Ross 308 chickens. In conclusion, using a slower-growing breed has beneficial effects on gut microbiota and fear responses of broilers, especially at slaughter age, whereas EF seems to have an impact only at an early stage of the life of broilers.
Collapse
Affiliation(s)
- Francesca Marcato
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, Netherlands
| | - Dirkjan Schokker
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, Netherlands
| | - Soumya Kanti Kar
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, Netherlands
| | - Alex Bossers
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
| | - Frank Harders
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, Netherlands
| | - Johanna M. J. Rebel
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, Netherlands
| | - Christine A. Jansen
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Elianne van der Valk
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, Netherlands
| | - Leo Kruijt
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, Netherlands
| | | | - Ingrid C. de Jong
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
5
|
Qu K, Shi M, Chen L, Liu Y, Yao X, Li X, Tan B, Xie S. Residual levels of dietary deltamethrin interfere with growth and intestinal health in Litopenaeus vannamei. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117376. [PMID: 39612679 DOI: 10.1016/j.ecoenv.2024.117376] [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: 05/05/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
To date, few study explored the damage of chronic dietary exposure to the lipophilic pesticide deltamethrin (DM) in aquatic animals, and it remains unclear whether its toxicity and residue levels would be affected by dietary lipid levels. Therefore, the present study aimed to elucidate the interactions between dietary lipid levels and DM levels in the Pacific white shrimp, focusing on growth performance, antioxidant capacity, and intestinal microbiota. DM has excellent insecticidal activity and has been used worldwide. Previous research has shown that environmental DM poses toxicity risks to aquatic animals. Six different diets were formulated to feed shrimp for 6 weeks with two lipid levels (6.96 %, 10.88 %) and three DM levels (0.2 mg·kg-1, 1 mg·kg-1, 5 mg·kg-1), namely LF0.2, LF1, LF5, HF0.2, HF1, HF5, respectively. Each diet was assigned to three net cages with a total of 18 cages (40 shrimp per tank, average weight (0.382±0.001 g), of which 0.2 mg·kg-1, are grouped in environmental DM control groups. The growth of shrimp was reduced as the dietary DM levels increased. When shrimp were fed a diet containing a high dose of DM, a reduction in their antioxidant capacity was also observed. Enzyme activity and gene expression related to lipid metabolism in hepatopancreas and hemolymph indicated a significant interaction between dietary lipid levels and DM in the lipid metabolism of shrimp. The terms of detoxification-related genes (gst, sult, cyp1a1) were upregulated in shrimp fed the high-dose DM. Additionally, the presence of DM in the diet severely harmed the hepatopancreas and intestinal histological morphology. DM in the diet increased the susceptibility of shrimp to pathogens and induced intestine microbiota dysbiosis, disrupting the balance of inter-species interactions. DM was not detected in the muscle and hepatopancreas of the shrimp after six weeks of exposure. In conclusion, the presence of DM in feed reduced the growth performance and antioxidant capacity of shrimp, damaging intestinal health. DM was rapidly metabolized by shrimp.
Collapse
Affiliation(s)
- Kangyuan Qu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Menglin Shi
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Liutong Chen
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yucheng Liu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xinzhou Yao
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoyue Li
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Beiping Tan
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China
| | - Shiwei Xie
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China.
| |
Collapse
|
6
|
Wang R, Patel D, Goruk S, Richard C, Field CJ. Feeding Docosahexaenoic Acid and Arachidonic Acid during Suckling and Weaning Contributes to Oral Tolerance Development by Beneficially Modulating the Intestinal Cytokine and Immunoglobulin Levels in an Allergy-Prone Brown Norway Rat Model. J Nutr 2024; 154:3790-3802. [PMID: 39401683 DOI: 10.1016/j.tjnut.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Suckling and weaning arachidonic acid (ARA) + docosahexaenoic acid (DHA) supplementation promoted oral tolerance (OT) development in pups, however, the effect of it on the intestine to promote OT development remains unknown. OBJECTIVE We aimed to explore the impact of this supplementation on intestinal fatty acid composition, structure, and indicators that are supportive of OT development. METHODS Allergy-prone Brown Norway dams were randomly assigned to a control (0% ARA, 0% DHA) or ARA + DHA diet (0.45% ARA, 0.8% DHA) during suckling (0-3 wk). At weaning (3-8 wk), offspring were randomly assigned to a control (0% ARA, 0% DHA) or ARA + DHA diet (0.5% ARA, 0.5% DHA). At 3 wk, offspring in each group received an oral gavage of sucrose or ovalbumin (OVA) solution for five consecutive days. At 7 wk, all offspring received an intraperitoneal OVA injection. At 8 wk, offspring were terminated to evaluate jejunum morphology and measure mucosal food allergy-related secretory immunoglobulin A (sIgA) and cytokines, ileum phospholipid and triglyceride fatty acid compositions, and fecal calprotectin. RESULTS Weaning ARA + DHA resulted in a higher percentage of DHA in ileum phospholipids and triglycerides (both P < 0.001), without affecting the percentage of ARA. Despite no lasting effect of suckling ARA + DHA on the DHA content in ileum phospholipids, a programming effect was found on the allergy-related intestinal immune profile [higher concentrations of mucosal IL-2 (P = 0.049) and sIgA (P = 0.033)]. OVA treatment resulted in a lower concentration of mucosal IL-6 (P = 0.026) regardless of dietary interventions. Offspring fed ARA + DHA during suckling and/or weaning had a higher concentration of mucosal transforming growth factor-beta (TGF-β) after OVA treatment but this was not observed in offspring fed control diets during suckling and weaning (P = 0.04). CONCLUSIONS Early life dietary ARA + DHA supplementation to allergy-prone rats enhanced the DHA concentration in intestinal phospholipids (weaning period) and increased the mucosal sIgA, IL-2, and TGF-β levels (suckling and weaning period), indicating its ability to create a tolerogenic intestinal environment to support OT development.
Collapse
Affiliation(s)
- Ren Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Dhruvesh Patel
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
Ali AQ, Sabir DK, Dawood AF, Abu-Rashed M, Hasari A, Gharqan F, Alnefaie S, Mohiddin LE, Tatry MM, Albadan DA, Alyami MM, Almutairi MF, Shawky LM. The potential liver injury induced by metronidazole-provoked disturbance of gut microbiota: modulatory effect of turmeric supplementation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9845-9858. [PMID: 38922353 DOI: 10.1007/s00210-024-03242-0] [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: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024]
Abstract
It has been reported that the gut-liver axis and intestinal microbiome contribute crucially to different liver diseases. So, targeting this hepato-intestinal connection may provide a novel treatment modality for hepatic disorders such as drug-induced liver injury (DILI). The present study thought to investigate the protective effect of turmeric (TUR) on metronidazole (MNZ)-induced liver damage and the possible association of the gut-liver axis and gut microbiota as a suggested underlying mechanism. In the first experiment, a MNZ-induced liver injury rat model was reproduced after 130 mg/kg oral MNZ administration for 30 days. Meanwhile, the treatment group was orally treated with 100 mg/kg turmeric daily. In the second experiment, fecal microbiome transplantation (FMT) was conducted, in which the fecal microbiome of each group in the first experiment was transplanted to a healthy corresponding group in the second experiment. The liver enzymes (aminotransferase (ALT) and aspartate aminotransferase (AST)) and histopathological examination were estimated to assess liver function. Inflammatory cytokines and oxidative markers were evaluated in the liver tissues. Histological analysis, intestinal barrier markers, and expression of tight junction proteins were measured for assessment of the intestinal injury. Changes in the gut microbial community and possible hepatic bacterial transmission were analyzed using 16S rRNA sequencing. MNZ induced intestinal and liver injuries which were significantly improved by turmeric. Increased firmicutes/bacteroidetes ratio and bacterial transmission due to gut barrier disruption were suggested. Moreover, TUR has maintained the gut microbial community by rebalancing and restoring bacterial proportions and abundance, thereby repairing the gut mucosal barrier and suppressing bacterial translocation. TUR protected against MNZ-induced gut barrier disruption. Reshaping of the intestinal bacterial composition and prohibition of the hepatic microbial translocation were suggested turmeric effects, potentially mitigating MNZ-related liver toxicity.
Collapse
Affiliation(s)
- Abdulaziz Qaid Ali
- Vision Colleges, Riyadh, Saudi Arabia.
- Faculty of Medicine, University of Sciences and Technology, Sana'a, Yemen.
| | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Amal F Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | - Lamiaa M Shawky
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
8
|
Oliveira ICCS, Marinsek GP, Gonçalves ARN, Lopes BS, Correia LVB, Da Silva RCB, Castro IB, Mari RB. Investigating tributyltin's toxic effects: Intestinal barrier and neuroenteric disruption in rat's jejunum. Neurotoxicology 2024; 105:208-215. [PMID: 39396746 DOI: 10.1016/j.neuro.2024.10.004] [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: 06/20/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The expansion of economic activities in coastal areas has significantly increased chemical contamination, leading to major environmental challenges. Contaminants enter the human body through the food chain, particularly via seafood and water consumption, triggering biomagnification and bioaccumulation processes. The gastrointestinal tract (GIT) acts as a selective barrier, protecting against chemical pollutants and maintaining homeostasis through a complex network of cells and immune responses. This study assessed impact of tributyltin (TBT), a highly toxic organometallic compound used in antifouling coatings for ships, on the GIT and myenteric neural plasticity in young rats. TBT exposure leads to histopathological changes, including epithelial detachment and inflammatory foci, especially at lower environmental doses. The study found that TBT causes significant reductions in villi height, increases in goblet cells and intraepithelial lymphocytes, and disrupts the myenteric plexus, with higher densities of extraganglionic neurons in exposed animals.
Collapse
Affiliation(s)
- I C C S Oliveira
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil.
| | - G P Marinsek
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - A R N Gonçalves
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - B S Lopes
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - L V B Correia
- UNIFESP, Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - R C B Da Silva
- UNIFESP, Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - I B Castro
- UNIFESP, Federal University of São Paulo, Institute of Marine Science, Baixada Santista Campus, Santos, SP, Brazil
| | - R B Mari
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| |
Collapse
|
9
|
Alwayli D, Jiang X, Liang J, Shah SRH, Ullah A, Abusidu MFZ, Shu W. Adjuvant Effect of Lactobacillus paracasei in Sublingual Immunotherapy of Asthmatic Mice. Pharmaceuticals (Basel) 2024; 17:1580. [PMID: 39770422 PMCID: PMC11678203 DOI: 10.3390/ph17121580] [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: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Sublingual immunotherapy (SLIT) has shown promise in mitigating allergic asthma symptoms; nevertheless, its high dose and prolonged duration of treatment raise safety concerns. This study explored the potential of Lactobacillus paracasei (L. paracasei) to enhance the effectiveness of SLIT in a mouse model of allergic asthma. Methods: Allergic asthma was induced in Balb/c mice following sensitization and challenge with a house dust mite (HDM) allergen. Subsequently, the mice were subjected to SLIT (66 and 132 µg) either alone or in combination with L. paracasei supplementation. Asthma-associated parameters, including rubbing frequency, IgE level, cytokine profiles, and histological changes, were evaluated to assess treatment efficacy. Results: mice that received SLIT 132 µg combined with the probiotic (combined 132) demonstrated a significant reduction in allergic symptoms (rubbing). This treatment strategy led to a marked IgE and eosinophil level decrease in serum; an increase in anti-inflammatory cytokines like IFN-γ and IL-10; and a reduction in pro-inflammatory cytokines IL-17 and TNF-α. The combination therapy also mitigated lung inflammation and supported the restoration of the structural integrity of the colon, promoting the recovery of goblet cells and mucus secretion. Probiotic treatment alone also effectively reduced IgE levels, increased IFN-γ, and decreased levels of IL-17 and TNF-α. Conclusions: The adjuvant effect of L. paracasei in enhancing SLIT represents a promising approach for improving asthma treatment efficacy.
Collapse
Affiliation(s)
- Dhafer Alwayli
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| | - Xiaoli Jiang
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| | - Jiaxu Liang
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| | - Syed Rafiq Hussain Shah
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| | - Atta Ullah
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| | - Mohammed F. Z. Abusidu
- Department of Biotechnology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China;
| | - Wen Shu
- Department of Pathogen Biology and Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (D.A.); (X.J.); (J.L.); (S.R.H.S.); (A.U.)
| |
Collapse
|
10
|
Alarfaj SJ, Bahaa MM, Elmasry TA, Elberri EI, El-Khateeb E, Hamouda AO, Salahuddin MM, Kamal M, Gadallah ANAA, Eltantawy N, Yasser M, Negm WA, Hamouda MA, Alsegiani AS, Alrubia S, Eldesoqui M, Abdallah MS. Fenofibrate as an Adjunct Therapy for Ulcerative Colitis: Targeting Inflammation via SIRT1, NLRP3, and AMPK Pathways: A Randomized Controlled Pilot Study. Drug Des Devel Ther 2024; 18:5239-5253. [PMID: 39575188 PMCID: PMC11578921 DOI: 10.2147/dddt.s490772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Background Ulcerative colitis (UC) is an idiopathic chronic inflammation of colonic and rectal mucosa. The peroxisome proliferator-activated receptor α (PPARα) has been identified as having protective effects in UC. Aim The study aimed to investigate the efficacy of fenofibrate, a PPARα agonist, in UC. Methods A total of 70 patients with mild to moderate UC were allocated randomly and assigned to two groups (n = 35 each) from Gastroenterology Department, Faculty of Medicine, Menoufia University. The mesalamine group received a placebo along with 1 g of mesalamine three times daily, while the fenofibrate group received 1 g of mesalamine three times and fenofibrate 160 mg once daily. The study duration was for six months. A gastroenterologist assessed patients by non-invasive Partial Mayo Score (PMS) and the Inflammatory Bowel Disease Questionnaire (IBDQ) to evaluate clinical response and remission. The serum levels of silent information regulator 1 (SIRT1), NOD-like receptor protein 3 (NLRP3), and adenosine monophosphate activated protein kinase (AMPK), as well as fecal calprotectin levels were examined to determine the biological effect of fenofibrate. Results After treatment, the fenofibrate group showed statistically significant reductions in PMS (p = 0.044) and improved digestive domain of IBDQ (p = 0.023). Additionally, there were significant decreases in serum NLRP3 (p = 0.041) and fecal calprotectin (p = 0.035), along with significant increases in SIRT1 (p = 0.002) and AMPK (p = 0.0003). The fenofibrate group also had higher response and remission rates compared to the mesalamine group. Conclusion Fenofibrate may be a promising adjunct for improving clinical outcomes, quality of life, and modulating inflammation in mild to moderate patients with UC. Trial Registration Identifier NCT05781698.
Collapse
Affiliation(s)
- Sumaiah J Alarfaj
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Thanaa A Elmasry
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman I Elberri
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Eman El-Khateeb
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amir O Hamouda
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Muhammed M Salahuddin
- Department of Biochemistry and Pharmacology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Marwa Kamal
- Department of Clinical Pharmacy, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | | | - Nashwa Eltantawy
- Department of Pharmacy Practice, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Mohamed Yasser
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Horus University, New Damietta, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, East Port Said National University, Port Said, Egypt
| | - Walaa A Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Manal A Hamouda
- Department of Clinical Pharmacy, Faculty of Pharmacy, Menofia University, Menofia, Egypt
| | - Amsha S Alsegiani
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sarah Alrubia
- Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud S Abdallah
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Sadat City (USC), Sadat City, Menoufia, Egypt
- Department of PharmD, Faculty of Pharmacy, Jadara University, Irbid, Jordan
| |
Collapse
|
11
|
Zubair M, Abouelnazar FA, Dawood AS, Pan J, Zheng X, Chen T, Liu P, Mao F, Yan Y, Chu Y. Microscopic messengers: microbiota-derived bacterial extracellular vesicles in inflammatory bowel disease. Front Microbiol 2024; 15:1481496. [PMID: 39606115 PMCID: PMC11600980 DOI: 10.3389/fmicb.2024.1481496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a persistent and complex condition accomplished by inflammation of the gastrointestinal system, encompassing Crohn's disease (CD) and ulcerative colitis (UC). This condition is caused by the combination of genetic predispositions, environmental triggers, and dysregulated immunological responses, which complicates diagnosis and treatment. The latest developments in gastroenterology have revealed the critical significance of the gut microbiota in the pathogenesis of IBD. Extracellular vesicles (EVs) are a type of microbial component that potentially regulate intestinal inflammation. The impact of microbiota-derived bacterial EVs (bEVs) on intestinal inflammation is mediated through several methods. They can intensify inflammation or stimulate defensive responses by delivering immunomodulatory cargo. Improved comprehension could enhance inventive diagnostic and treatment strategies for IBD. This study aimed to explore the relationship between microbiota-derived bEVs and the complex nature of IBD. We performed a thorough analysis of the formation, composition, mechanisms of action, diagnostic possibilities, therapeutic implications, and future prospects of these microbiota-derived bEVs.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fatma A. Abouelnazar
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
- Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | - Ali Sobhy Dawood
- Medicine and Infectious Diseases Department, Faculty of Veterinary Medicine, University of Sadat City, Sadat, Egypt
| | - Jingyun Pan
- Department of Traditional Chinese Medicine, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Xuwen Zheng
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Tao Chen
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Pengjun Liu
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Ying Chu
- Wujin Clinical College, Xuzhou Medical University, Changzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Zhang J, Chen Y, Guo X, Li X, Zhang R, Wang M, Zhu W, Yu K. The gut microbial metabolite indole-3-aldehyde alleviates impaired intestinal development by promoting intestinal stem cell expansion in weaned piglets. J Anim Sci Biotechnol 2024; 15:150. [PMID: 39511673 PMCID: PMC11545576 DOI: 10.1186/s40104-024-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Weaning stress-induced diarrhea is widely recognized as being associated with gut microbiota dysbiosis. However, it has been challenging to clarify which specific intestinal microbiota and their metabolites play a crucial role in the antidiarrhea process of weaned piglets. RESULTS In this study, we first observed that piglets with diarrhea exhibited a lower average daily gain and higher diarrhea score, and elevated levels of lipopolysaccharide (LPS) and D-lactate (D-LA) compared to healthy piglets. Subsequently, we analyzed the differences in intestinal microbial composition and metabolite levels between healthy and diarrheal weaned piglets. Diarrheal piglets demonstrated intestinal microbiota dysbiosis, characterized primarily by a higher Firmicutes to Bacteroidota ratio, a deficiency of Lactobacillus amylovorus and Lactobacillus reuteri, and an increased abundance of Bacteroides sp.HF-5287 and Bacteroides thetaiotaomicron. Functional profiling of the gut microbiota based on Kyoto Encyclopedia of Genes and Genomes (KEGG) data was performed, and the results showed that tryptophan metabolism was the most significantly inhibited pathway in piglets with diarrhea. Most tryptophan metabolites were detected at lower concentrations in diarrheal piglets than in healthy piglets. Furthermore, we explored the effects of dietary indole-3-aldehyde (IAld), a key tryptophan metabolite, on intestinal development and gut barrier function in weaned piglets. Supplementation with 100 mg/kg IAld in the diet increased the small intestine index and improved intestinal barrier function by promoting intestinal stem cell (ISC) expansion in piglets. The promotion of ISC expansion by IAld was also confirmed in porcine intestinal organoids. CONCLUSIONS These findings revealed that intestinal microbial tryptophan metabolite IAld alleviates impaired intestinal development by promoting ISC expansion in weaned piglets.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yahui Chen
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xin Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruofan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
- Wujiang Animal Health Inspection Institute, Suzhou, 215200, China
| | - Mengting Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
13
|
Zhang Y, Song M, Fan J, Guo X, Tao S. Impact of probiotics-derived extracellular vesicles on livestock gut barrier function. J Anim Sci Biotechnol 2024; 15:149. [PMID: 39506860 PMCID: PMC11542448 DOI: 10.1186/s40104-024-01102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/05/2024] [Indexed: 11/08/2024] Open
Abstract
Probiotic extracellular vesicles (pEVs) are biologically active nanoparticle structures that can regulate the intestinal tract through direct or indirect mechanisms. They enhance the intestinal barrier function in livestock and poultry and help alleviate intestinal diseases. The specific effects of pEVs depend on their internal functional components, including nucleic acids, proteins, lipids, and other substances. This paper presents a narrative review of the impact of pEVs on the intestinal barrier across various segments of the intestinal tract, exploring their mechanisms of action while highlighting the limitations of current research. Investigating the mechanisms through which probiotics operate via pEVs could deepen our understanding and provide a theoretical foundation for their application in livestock production.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Mengzhen Song
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Xuming Guo
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China.
| |
Collapse
|
14
|
Abeltino A, Hatem D, Serantoni C, Riente A, De Giulio MM, De Spirito M, De Maio F, Maulucci G. Unraveling the Gut Microbiota: Implications for Precision Nutrition and Personalized Medicine. Nutrients 2024; 16:3806. [PMID: 39599593 PMCID: PMC11597134 DOI: 10.3390/nu16223806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Recent studies have shown a growing interest in the complex relationship between the human gut microbiota, metabolism, and overall health. This review aims to explore the gut microbiota-host association, focusing on its implications for precision nutrition and personalized medicine. The objective is to highlight how gut microbiota modulate metabolic and immune functions, contributing to disease susceptibility and wellbeing. The review synthesizes recent research findings, analyzing key studies on the influence of gut microbiota on lipid and carbohydrate metabolism, intestinal health, neurobehavioral regulation, and endocrine signaling. Data were drawn from both experimental and clinical trials examining microbiota-host interactions relevant to precision nutrition. Our findings highlight the essential role of gut microbiota-derived metabolites in regulating host metabolism, including lipid and glucose pathways. These metabolites have been found to influence immune responses and gut barrier integrity. Additionally, the microbiota impacts broader physiological processes, including neuroendocrine regulation, which could be crucial for dietary interventions. Therefore, understanding the molecular mechanisms of dietary-microbiota-host interactions is pivotal for advancing personalized nutrition strategies. Tailored dietary recommendations based on individual gut microbiota compositions hold promise for improving health outcomes, potentially revolutionizing future healthcare approaches across diverse populations.
Collapse
Affiliation(s)
- Alessio Abeltino
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Duaa Hatem
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Cassandra Serantoni
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Alessia Riente
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Michele Maria De Giulio
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Marco De Spirito
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Flavio De Maio
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Maulucci
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| |
Collapse
|
15
|
Vieco-Saiz N, Prévéraud DP, Pinloche E, Morat A, Govindin P, Blottière HM, Matthieu E, Devillard E, Consuegra J. Unraveling the benefits of Bacillus subtilis DSM 29784 poultry probiotic through its secreted metabolites: an in vitro approach. Microbiol Spectr 2024; 12:e0017724. [PMID: 39287453 PMCID: PMC11537077 DOI: 10.1128/spectrum.00177-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/04/2024] [Indexed: 09/19/2024] Open
Abstract
The probiotic Bacillus subtilis 29784 (Bs29784) sustains chicken's intestinal health, enhancing animal resilience and performance through the production of the bioactive metabolites hypoxanthine (HPX), niacin (NIA), and pantothenate (PTH). Here, using enterocyte in vitro models, we determine the functional link between these metabolites and the three pillars of intestinal resilience: immune response, intestinal barrier, and microbiota. We evaluated in vitro the capacity of Bs29784 vegetative cells, spores, and metabolites to modulate global immune regulators (using HT-29-NF-κB and HT-29-AP-1 reporter cells), intestinal integrity (HT-29-MUC2 reporter cells and Caco-2 cells), and cytokine production (Caco-2 cells). Finally, we simulated intestinal fermentations using chicken's intestinal contents as inocula to determine the effect of Bs29784 metabolites on the microbiota and their fermentation profile. Bs29784 vegetative cells reduced the inflammatory response more effectively than spores, indicating that their benefit is linked to metabolic activity. To assess this hypothesis, we studied Bs29784 metabolites individually. The results showed that each metabolite had different beneficial effects. PTH and NIA reduced the activation of the pro-inflammatory pathways AP-1 and NF-κB. HPX upregulated mucin production by enhancing MUC2 expression. HPX, NIA, and PTH increased cell proliferation. PTH and HPX increased epithelial resilience to an inflammatory challenge by limiting permeability increase. In cecal fermentations, NIA increased acetate, HPX increased butyrate, whereas PTH increased acetate, butyrate, and propionate. In ileal fermentations, PTH increased butyrate. All molecules modulated microbiota, explaining the different fermentation patterns. Altogether, we show that Bs29784 influences intestinal health by acting on the three lines of resilience via its secreted metabolites. IMPORTANCE Probiotics provide beneficial metabolites to its host. Here, we describe the mode of action of a commonly used probiotic in poultry, Bs29784. By using in vitro cellular techniques and simulated chickens' intestinal model, we show the functional link between Bs29784 metabolites and the three lines of animal resilience. Indeed, both Bs29784 vegetative cells and its metabolites stimulate cellular anti-inflammatory responses, strengthen intestinal barrier, and positively modulate microbiota composition and fermentative profile. Taken together, these results strengthen our understanding of the effect of Bs29784 on its host and explain, at least partly, its positive effects on animal health, resilience, and performance.
Collapse
Affiliation(s)
- Nuria Vieco-Saiz
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | | | - Eric Pinloche
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | - Aurélien Morat
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Pauline Govindin
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Hervé M. Blottière
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
- Nantes Université, INRAE, UMR 1280, PhAN, Nantes, France
| | - Elliot Matthieu
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Estelle Devillard
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | - Jessika Consuegra
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| |
Collapse
|
16
|
Elkhaiat IA, El-Kassas S, Abdo SE, El-Naggar K, Shalaby HK, Nofal RY, Farag MR, Azzam MM, Lestingi A. Leverage of lysozyme dietary supplementation on gut health, hematological, antioxidant, and immune parameters in different plumage-colors Japanese quails. Poult Sci 2024; 104:104474. [PMID: 39571202 PMCID: PMC11617721 DOI: 10.1016/j.psj.2024.104474] [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: 08/21/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 12/08/2024] Open
Abstract
The current study was conducted on two different feather-colored Japanese quail varieties (brown and white) to examine the impact of lysozyme (LZ) dietary supplementation on growth performance, hematological profile, serum lysozyme, phagocytic and antioxidant activities, along with the gut status and the relative expression of some antioxidant- and immune-related genes. Two forms of LZ; extracted from egg white (natural LZ (NLZ)), and the commercial LZ (CLZ) were included in this experiment. For each quail variety, 240 birds were randomly allocated into four groups with four replicates per group. The first group (control) ate the basal diet (BD) only. The other groups ate the BD supplemented with commercial lysozyme (CLZ, at 100 mg/kg diet), NLZ at 100 (NLZ1) and 200 (NLZ2) mg/kg diet. Different LZ treatments differentially modulated the quail's growth performance with significant increases in the final body weight of white-feathered quails fed the NLZ1 compared to other treatments. The NLZ2 and CLZ noticeably increased the total antioxidant activity (TA) in the white- and brown-feathered quails, respectively. Also, all LZ groups displayed distinct increases in the serum lysozyme and phagocytic activities. For gut status, both varieties exhibited increases in intestinal villi length and goblet cell count with significant reductions in the total lactobacillus, total coliform, and total bacterial counts. These effects were linked with marked modulations of SOD, CAT, GPX, andIL-1βgene expression levels in both quail varieties. Therefore, the LZ could differentially impact quail growth, immune and antioxidant status as well as gut health.
Collapse
Affiliation(s)
- Ibrahim A Elkhaiat
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Seham El-Kassas
- Animal, Poultry and Fish Breeding and Production, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Safaa E Abdo
- Genetics and Genetic Engineering, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Karima El-Naggar
- Department of Nutrition and Veterinary Clinical Nutrition, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Haitham K Shalaby
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Reyad Y Nofal
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Mayada R Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt.
| | - Mahmoud M Azzam
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Antonia Lestingi
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km 3, 70010, Valenzano, BA, Italy
| |
Collapse
|
17
|
Ma WW, Huang ZQ, Liu K, Li DZ, Mo TL, Liu Q. The role of intestinal microbiota and metabolites in intestinal inflammation. Microbiol Res 2024; 288:127838. [PMID: 39153466 DOI: 10.1016/j.micres.2024.127838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/11/2024] [Accepted: 07/13/2024] [Indexed: 08/19/2024]
Abstract
With the imbalance of intestinal microbiota, the body will then face an inflammatory response, which has serious implications for human health. Bodily allergies, injury or pathogens infections can trigger or promote inflammation and alter the intestinal environment. Meanwhile, excessive changes in the intestinal environment cause the imbalance of microbial homeostasis, which leads to the proliferation and colonization of opportunistic pathogens, invasion of the body's immune system, and the intensification of inflammation. Some natural compounds and gut microbiota and metabolites can reduce inflammation; however, the details of how they interact with the gut immune system and reduce the gut inflammatory response still need to be fully understood. The review focuses on inflammation and intestinal microbiota imbalance caused by pathogens. The body reacts differently to different types of pathogenic bacteria, and the ingestion of pathogens leads to inflamed gastrointestinal tract disorders or intestinal inflammation. In this paper, unraveling the interactions between the inflammation, pathogenic bacteria, and intestinal microbiota based on inflammation caused by several common pathogens. Finally, we summarize the effects of intestinal metabolites and natural anti-inflammatory substances on inflammation to provide help for related research of intestinal inflammation caused by pathogenic bacteria.
Collapse
Affiliation(s)
- Wen-Wen Ma
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Zhi-Qiang Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Kun Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - De-Zhi Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China
| | - Tian-Lu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| | - Qing Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China.
| |
Collapse
|
18
|
Manithody C, Denton C, Mehta S, Carter J, Kurashima K, Bagwe A, Swiderska-Syn M, Guzman M, Besmer S, Jain S, McHale M, Qureshi K, Nazzal M, Caliskan Y, Long J, Lin CJ, Hutchinson C, Ericsson AC, Jain AK. Intraduodenal fecal microbiota transplantation ameliorates gut atrophy and cholestasis in a novel parenteral nutrition piglet model. Am J Physiol Gastrointest Liver Physiol 2024; 327:G640-G654. [PMID: 39163019 PMCID: PMC11559648 DOI: 10.1152/ajpgi.00012.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Total parenteral nutrition (TPN) provides lifesaving nutritional support intravenously; however, it is associated with significant side effects. Given gut microbial alterations noted with TPN, we hypothesized that transferring fecal microbiota from healthy controls would restore gut-systemic signaling in TPN and mitigate injury. Using our novel ambulatory model (US Patent: US 63/136,165), 31 piglets were randomly allocated to enteral nutrition (EN), TPN only, TPN + antibiotics (TPN-A), or TPN + intraduodenal fecal microbiota transplant (TPN + FMT) for 14 days. Gut, liver, and serum were assessed through histology, biochemistry, and qPCR. Stool samples underwent 16 s rRNA sequencing. Permutational multivariate analysis of variance, Jaccard, and Bray-Curtis metrics were performed. Significant bilirubin elevation in TPN and TPN-A versus EN (P < 0.0001) was prevented with FMT. IFN-G, TNF-α, IL-β, IL-8, and lipopolysaccharide (LPS) were significantly higher in TPN (P = 0.009, P = 0.001, P = 0.043, P = 0.011, P < 0.0001), with preservation upon FMT. Significant gut atrophy by villous-to-crypt ratio in TPN (P < 0.0001) and TPN-A (P = 0.0001) versus EN was prevented by FMT (P = 0.426 vs. EN). Microbiota profiles using principal coordinate analysis demonstrated significant FMT and EN overlap, with the largest separation in TPN-A followed by TPN, driven primarily by Firmicutes and Fusobacteria. TPN-altered gut barrier was preserved upon FMT; upregulated cholesterol 7 α-hydroxylase and bile salt export pump in TPN and TPN-A and downregulated fibroblast growth factor receptor 4, EGF, farnesoid X receptor, and Takeda G Protein-coupled Receptor 5 (TGR5) versus EN was prevented by FMT. This study provides novel evidence of prevention of gut atrophy, liver injury, and microbial dysbiosis with intraduodenal FMT, challenging current paradigms into TPN injury mechanisms and underscores the importance of gut microbes as prime targets for therapeutics and drug discovery.NEW & NOTEWORTHY Intraduodenal fecal microbiota transplantation presents a novel strategy to mitigate complications associated with total parenteral nutrition (TPN), highlighting gut microbiota as a prime target for therapeutic and diagnostic approaches. These results from a highly translatable model provide hope for TPN side effect mitigation for thousands of chronically TPN-dependent patients.
Collapse
Affiliation(s)
- Chandrashekhara Manithody
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Christine Denton
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Shaurya Mehta
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Jasmine Carter
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Kento Kurashima
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Ashlesha Bagwe
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Marzena Swiderska-Syn
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Miguel Guzman
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Sherri Besmer
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Sonali Jain
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Matthew McHale
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Kamran Qureshi
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Mustafa Nazzal
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Yasar Caliskan
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - John Long
- Department of Comparative Medicine, Saint Louis University, Saint Louis, Missouri, United States
| | - Chien-Jung Lin
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Chelsea Hutchinson
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States
| | - Ajay Kumar Jain
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, United States
| |
Collapse
|
19
|
Song HA, Jang SY, Park MJ, Kim SW, Kang CG, Lee JH, Kim HJ, Kim J, Lee JK, Chung KS, Lee KT. Immunostimulation Signaling via Toll-like Receptor 2 Activation: A Molecular Mechanism of Lactococcus lactis OTG1204 In Vitro and In Vivo. Nutrients 2024; 16:3629. [PMID: 39519462 PMCID: PMC11547582 DOI: 10.3390/nu16213629] [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: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION The immune system's defense against pathogens involves innate and adaptive responses, crucial in maintaining overall health. Immunosuppressed states render individuals more susceptible to potential diseases, indicating the need for effective strategies to bolster immune functions. OBJECTIVES Although the immunostimulatory effects of various probiotics have been studied, the specific effects and molecular mechanisms of Lactococcus lactis OTG1204 (OTG1204) remain unknown. In this study, the aim was to investigate the molecular mechanisms of OTG1204 in RAW 264.7 macrophages, the key effector cells of the innate immune system involved in host defense and inflammatory responses. Additionally, in this study, the effects of OTG1204 on cyclophosphamide (CTX)-induced immunosuppression states were investigated, thereby demonstrating its potential as an immune stimulant. METHODS To assess the macrophage activation ability and underlying mechanisms of OTG1204, RAW 264.7 cells were utilized with transfection, enzyme-linked immunosorbent assay, and quantitative real-time PCR analyses. Furthermore, to evaluate the immunostimulatory effects under immunosuppressed conditions, CTX-induced immunosuppression mice model was employed, and analyses were performed using hematoxylin and eosin staining, flow cytometry, and microbiota examination. RESULTS OTG1204 activated RAW 264.7 macrophages, leading to increased production of nitric oxide, prostaglandin E2, and cytokines. This immune activation was mediated through the upregulation of toll-like receptor 2, which subsequently activated the nuclear factor-κB (NF-kB) and mitogen-activated protein kinase (MAPK)/activator protein 1 (AP-1) pathways, thereby stimulating the immune response. In CTX-treated mice, OTG1204 recovered body weight, spleen, and mesenteric lymph node indices, and natural killer cell activity. It re-established populations of innate and adaptive immune cells and activated T cells to secrete cytokines. We also examined the gut barrier integrity and microbiota composition to assess OTG1204's impact on intestinal health, as these factors play a significant role in immune enhancement. OTG1204 enhanced gut barrier integrity by upregulating mucin 2 and tight junction proteins and modulated the gut microbiota by restoring the Firmicutes/Bacteroidetes balance and reducing the abundance of Actinobacteria and Tenericutes. CONCLUSION These results suggest that OTG1204 may serve as an effective probiotic for immune enhancement and gut health management by targeting the NF-κB and MAPK/AP-1 pathways, with minimal side effects.
Collapse
Affiliation(s)
- Hyeon-A Song
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seo-Yun Jang
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Min-Ji Park
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung Wook Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Choon Gil Kang
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Joo Hyun Lee
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Hye-Jin Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jiheon Kim
- Ottogi Research Center, Anyang 14060, Republic of Korea; (S.W.K.); (C.G.K.); (J.H.L.); (H.-J.K.); (J.K.)
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-A.S.); (S.-Y.J.); (M.-J.P.); (K.-S.C.)
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
20
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619652. [PMID: 39484519 PMCID: PMC11526895 DOI: 10.1101/2024.10.22.619652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal epithelial barrier dysfunction, and aberrant inflammatory responses. In addition, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, reduction in the abundance of beneficial commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no FDA-approved countermeasures that can treat radiation-induced GI injury. To meet this critical need, Synedgen Inc., has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy total body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory response mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera, while suppressing potentially pathogenic bacteria compared with vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and effectively reducing proinflammatory responses. Further development of this drug as an FDA-approved medical countermeasure will be of critical importance in the event of a radiation public health emergency.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
21
|
Wardenaar FC, Chan Y, Clear AM, Schott K, Mohr AE, Ortega-Santos CP, Seltzer RGN, Pugh J. The Gastrointestinal Symptom Rating Scale has a Good Test-Retest Reliability in Well-Trained Athletes With and Without Previously Self-Identified Gastrointestinal Complaints. Sports Med 2024:10.1007/s40279-024-02122-0. [PMID: 39363029 DOI: 10.1007/s40279-024-02122-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Athletes often report gastrointestinal (GI) complaints. Standardized validated tests validated in athletes are lacking. OBJECTIVE The objective of the current study was to investigate the test-retest reliability of the gastrointestinal symptoms rating scale (GSRS), a disease-specific instrument of 15 items to quantify the severity of various GI symptoms. METHODS For this purpose, a 3-week repeated measurements design was used. The mean difference (Wilcoxon signed rank test), associations (Spearman correlations), and systematic difference using Bland-Altman calculations for repeated measurements, as well as its internal consistency (Cronbach's alpha) on testing day 1 and day 2 were analyzed, with significance set at p ≤ 0.05. A total of n = 70 well-trained athletes (26.1 ± 5.4 years, of which 40% were female) were included. RESULTS A high Cronbach's α for GSRS was found on testing day 1 (0.825), and day 2 (0.823), suggesting a good and comparable internal consistency of the questionnaire. When assessing the multilevel temporal stability for total GSRS scores (28.0, IQR 22.0-36.3 vs 26.5, IQR 18.0-35.0), there was a small but significant difference (Z = - 2.489, and p = 0.013), but a fair correlation between day scores (r = 0.68, p < 0.001), and a Bland-Altman reporting difference between questionnaires within 10% of the total GSRS score, without significant reporting bias (p = 0.38). In most cases, except for hunger, burping, and loose stools, the individual GSRS items were reported in line with total scores and similar for sex. CONCLUSION In conclusion, the GSRS is reliable when used with athletes, with good internal consistency for most symptoms independently of sex, except for hunger, burping, and loose stools.
Collapse
Affiliation(s)
- Floris C Wardenaar
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA.
| | - Yat Chan
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Anna Marie Clear
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Kinta Schott
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Center for Health Through Microbiomes, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Carmen P Ortega-Santos
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Ryan G N Seltzer
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Jamie Pugh
- School of Sport and Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, Liverpool, UK
| |
Collapse
|
22
|
Lee J, Lee MS, Kim Y. Effects of Green Tea and Java Pepper Mixture on Gut Microbiome and Colonic MicroRNA-221/222 in Mice with Dextran Sulfate Sodium-Induced Colitis. Prev Nutr Food Sci 2024; 29:279-287. [PMID: 39371512 PMCID: PMC11450278 DOI: 10.3746/pnf.2024.29.3.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
In this study, we aimed to investigate the regulatory effects of a green tea and java pepper mixture (GTP) on the gut microbiome and microRNA (miR)-221/222 expression in mice with dextran sulfate sodium (DSS)-induced colitis. Male C57BL/6J mice were divided into four groups: DSS-, DSS+, GTP50, and GTP100. In the GTP50 and GTP100 groups, GTP was orally administered to mice at doses of 50 and 100 mg/kg body weight, respectively, every day for 2 weeks, and colitis was induced in the DSS+, GTP50, and GTP100 groups by adding 3% DSS to their drinking water for 1 week. GTP was found to mitigate the severity of inflammation and the damage to goblet cells caused by DSS-induced colitis. The results showed that compared with the DSS- group, the relative abundance of Bacteroidetes was increased and that of Proteobacteria and Candidatus Melainabacteria was decreased in the GTP100 group. The ratio of Firmicutes to Bacteroidetes was also reduced in the GTP100 group. However, GTP administration did not modulate the microbial diversity. GTP administration upregulated the mRNA and protein levels of occludin and zonula occludens 1. In addition, GTP effectively downregulated the expression of miR-221 and miR-222. Overall, GTP altered the gut microbiota composition and downregulated colonic miR-221/222 expression in mice with DSS-induced colitis.
Collapse
Affiliation(s)
- Jumi Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Mak-Soon Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yangha Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
23
|
Hao L, Yan Y, Huang G, Li H. From gut to bone: deciphering the impact of gut microbiota on osteoporosis pathogenesis and management. Front Cell Infect Microbiol 2024; 14:1416739. [PMID: 39386168 PMCID: PMC11461468 DOI: 10.3389/fcimb.2024.1416739] [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: 04/13/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Osteoporosis (OP) is characterized by decreased bone mineral density (BMD) and increased fracture risk, poses a significant global health burden. Recent research has shed light on the bidirectional relationship between gut microbiota (GM) and bone health, presenting a novel avenue for understanding OP pathogenesis and developing targeted therapeutic interventions. This review provides a comprehensive overview of the GM-bone axis, exploring the impact of GM on OP development and management. We elucidate established risk factors and pathogenesis of OP, delve into the diversity and functional changes of GM in OP. Furthermore, we examine experimental evidence and clinical observations linking alterations in GM composition or function with variations in BMD and fracture risk. Mechanistic insights into microbial mediators of bone health, such as microbial metabolites and products, are discussed. Therapeutic implications, including GM-targeted interventions and dietary strategies, are also explored. Finally, we identify future research directions and challenges in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Linjie Hao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yuzhu Yan
- Clinical Laboratory of Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Guilin Huang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hui Li
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
24
|
Gao YY, Zhou YH, Liu XP, Di B, He JY, Wang YT, Guo PT, Zhang J, Wang CK, Jin L. Ganoderma lucidum polysaccharide promotes broiler health by regulating lipid metabolism, antioxidants, and intestinal microflora. Int J Biol Macromol 2024; 280:135918. [PMID: 39322164 DOI: 10.1016/j.ijbiomac.2024.135918] [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: 04/01/2024] [Revised: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Ganoderma lucidum polysaccharides (GLP) are the primary bioactive macromolecular compounds of Ganoderma lucidum, possessing antioxidant and immunomodulatory effects. Hot water extract of Juncao-substrate Ganoderma Lucidum residue (HWE-JGLR) is abundant in GLP. There are few research reports on the application of HWE-JGLR in animal husbandry. Therefore, this study aims to investigate the effects of HWE-JGLR supplementation on growth performance, serum biochemistry, the antioxidant function of serum and liver, and the intestinal microbiota of yellow-feathered broilers. The control group was fed a corn-soybean meal basal diet, while the HJ I, II, and III groups received diets supplemented with 0.25 %, 0.5 %, and 1 % of HWE-JGLR, respectively. Results showed that HWE-JGLR increased the serum HDL-C content and decreased the TG content in broilers. Moreover, HWE-JGLR enhanced the antioxidant function by the Keap1-Nrf2/ARE signaling pathway and the antioxidative enzyme in broilers. In addition, the cecum of the metagenomic analysis of 16S rRNA showed that the relative abundance of no-rank Ruminococcacea was increased in the HJ I group. Our findings indicate that HWE-JGLR has strong potential for development as a green feed additive based on its functions of lipid-lowering, antioxidation, and the modulation of gut microbiota composition.
Collapse
Affiliation(s)
- Yu-Yun Gao
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ying-Huan Zhou
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiao-Ping Liu
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Bin Di
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jia-Yi He
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ya-Ting Wang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ping-Ting Guo
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing Zhang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chang-Kang Wang
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ling Jin
- China National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
25
|
van Dinteren S, Araya-Cloutier C, Bastiaan-Net S, Boudewijn A, van Heek T, Vincken JP, Witkamp R, Meijerink J. Biotransformation and Epithelial Toxicity of Prenylated Phenolics from Licorice Roots ( Glycyrrhiza spp.) in 3D Apical-Out Mucus-Producing Human Enteroids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20396-20409. [PMID: 39240776 PMCID: PMC11421016 DOI: 10.1021/acs.jafc.4c03120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 09/08/2024]
Abstract
Apical-out enteroids mimic the in vivo environment well due to their accessible apical surface and mucus layer, making them an ideal model for studying the impact of (bioactive) food compounds. Generated human ileal apical-out enteroids showed a fucose-containing mucus layer surrounding the apical brush border on their exposure side, indicating their physiological relevance. Effects on the mucosal epithelium of antibacterial prenylated phenolics (glabridin, licochalcone A, and glycycoumarin) from licorice roots were investigated for cytotoxicity, cell viability, barrier integrity, and biotransformation. At concentrations up to 500 μg mL-1, licochalcone A and glycycoumarin did not significantly affect apical-out enteroids, with cytotoxicities of -6 ± 2 and -2 ± 2% and cell viabilities of 77 ± 22 and 77 ± 13%, respectively (p > 0.05). Conversely, 500 μg mL-1 glabridin induced significant cytotoxicity (31 ± 25%, p < 0.05) and reduced cell viability (21 ± 14%, p < 0.01). Apical-out enteroids revealed differential sensitivities to prenylated phenolics not observed in apical-in enteroids and Caco-2 cells. Both enteroid models showed phase II biotransformation but differed in the extent of glucuronide conversion. The apical mucus layer of apical-out enteroids likely contributed to these differential interactions, potentially due to differences in electrostatic repulsion. This study underscores the relevance of 3D apical-out enteroid models and highlights the promise of prenylated phenolics for antimicrobial applications.
Collapse
Affiliation(s)
- Sarah van Dinteren
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Carla Araya-Cloutier
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen
Food & Biobased Research, Wageningen
University & Research, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Anouk Boudewijn
- Wageningen
Food & Biobased Research, Wageningen
University & Research, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Tjarda van Heek
- Department
of Abdominal Surgery, Hospital Gelderse
Vallei, Willy Brandtlaan 10, Ede 6716 RP, The Netherlands
| | - Jean-Paul Vincken
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Renger Witkamp
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Jocelijn Meijerink
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| |
Collapse
|
26
|
Chen X, Gasaly N, Tang X, Walvoort MT, de Vos P. The effect of nerve cells on the intestinal barrier function and the influence of human milk oligosaccharides (hMOs) on the intestinal neuro-epithelial crosstalk. Curr Res Food Sci 2024; 9:100851. [PMID: 39314222 PMCID: PMC11417580 DOI: 10.1016/j.crfs.2024.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
The intestinal epithelium is an important gatekeeper of the human body by forming a barrier for the luminal content of the intestine. The barrier function is regulated by a complex crosstalk between different cell types, including cells from the enteric nervous system (ENS). ENS is considered to influence gastrointestinal processes and functions, but its direct effect on epithelial barrier function remains to be confirmed. To investigate the effect of nerve cells on the gut barrier function, an in vitro co-culture system was established in which T84 intestinal epithelial cells and SH-SY5Y nerve cells were seeded in ratios of 29:1 and 14:1. When the epithelial barrier was disrupted with the calcium ionophores A23187, we found that nerve cells exert a protective effect on A23187-induced disruption and that this protective effect is nerve cell concentration-dependent. This was demonstrated by rescuing effects on transepithelial electrical resistance (TEER) and upregulation of tight junction (TJ) protein expression. Furthermore, we studied whether similar rescuing effects could be achieved with the human milk oligosaccharides (hMOs) 2'-fucosyllactose (2'-FL) and 3-fucosyllactose (3-FL). Our results illustrate that in the presence of nerve cells 2'-FL and 3-FL do not have any additional rescuing effects, but that these hMOs can substitute the rescuing effects of nerve cells in the absence of nerve cells. Meanwhile, 2'-FL and 3-FL show different regulation effects on TJ expression. These findings provide valuable insights into potential therapeutic strategies for maintaining intestinal barrier integrity.
Collapse
Affiliation(s)
- Xiaochen Chen
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Naschla Gasaly
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Xin Tang
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marthe T.C. Walvoort
- Stratingh Institute for Chemistry, Department of Chemical Biology, University of Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Immunoendocrinology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
27
|
Cui Y, Zhang M, Wang H, Yu T, Zhang A, Lin G, Guo Y, Wu Y. Organic Trace Minerals Enhance the Gut Health of British Shorthair Cats by Regulating the Structure of Intestinal Microbiota. Metabolites 2024; 14:494. [PMID: 39330501 PMCID: PMC11434296 DOI: 10.3390/metabo14090494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Trace minerals are essential for biological processes, including enzyme function, immune response, and hormone synthesis. The study assessed the effects of different dietary trace minerals on the gut health, microbiota composition, and immune function of cats. Eighteen adult British Shorthair cats were divided into three groups receiving inorganic trace minerals (ITM), a 50/50 mix of inorganic and organic trace minerals (ITM + OTM), or organic trace minerals (OTM) for 28 days. The OTM showed enhanced immune capacities, reduced intestinal barrier function, and lower inflammation condition. The OTM altered gut microbiota diversity, with a lower Simpson index and higher Shannon index (p < 0.05). Specifically, the abundance of Bacteroidota, Lachnospiraceae, and Prevotella in the OTM group were higher than the ITM group (p < 0.05). Metabolomic analysis identified 504 differential metabolites between the OTM and ITM groups (p < 0.05, VIP-pred-OPLS-DA > 1), affecting pathways related to steroid hormone biosynthesis and glycerophospholipid metabolism (p < 0.05, VIP-pred-OPLS-DA > 2). Additionally, there was a significant correlation between intestinal microbiota and differential metabolites. To conclude, dietary OTM can modulate the gut metabolite and microbiota composition, enhance immune and intestinal barrier function, and mitigate inflammation in cats, highlighting the benefit of using OTM in feline diet to promote the intestinal and overall health.
Collapse
Affiliation(s)
- Yingyue Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Tong Yu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Anxuan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Gang Lin
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (G.L.); (Y.G.)
| | - Yuhan Guo
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (G.L.); (Y.G.)
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| |
Collapse
|
28
|
Kanika NH, Hou X, Liu H, Dong Y, Wang J, Wang C. Specific gut microbiome's role in skin pigmentation: insights from SCARB1 mutants in Oujiang colour common carp. J Appl Microbiol 2024; 135:lxae226. [PMID: 39243120 DOI: 10.1093/jambio/lxae226] [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: 12/06/2023] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024]
Abstract
AIMS Beyond the pivotal roles of the gut microbiome in initiating physiological processes and modulating genetic factors, a query persists: Can a single gene mutation alter the abundance of the gut microbiome community? Not only this, but the intricate impact of gut microbiome composition on skin pigmentation has been largely unexplored. METHODS AND RESULTS Based on these premises, our study examines the abundance of lipase-producing gut microbes about differential gene expression associated with bile acid synthesis and lipid metabolism-related blood metabolites in red (whole wild) and white (whole white wild and SCARB1-/- mutant) Oujiang colour common carp. Following the disruption of the SCARB1 gene in the resulting mutant fish with white body colour (SCARB1-/-), there is a notable decrease in the abundance of gut microbiomes (Bacillus, Staphylococcus, Pseudomonas, and Serratia) associated with lipase production. This reduction parallels the downregulation seen in wild-type white body colour fish (WW), as contrasting to the wild-type red body colour fish (WR). Meanwhile, in SCARB1-/- fish, there was a downregulation noted not only at the genetic and metabolic levels but also a decrease in lipase-producing bacteria. This consistency with WW contrasts significantly with WR. Similarly, genes involved in the bile acid synthesis pathway, along with blood metabolites related to lipid metabolism, exhibited downregulation in SCARB1-/- fish. CONCLUSIONS The SCARB1 knockout gene blockage led to significant alterations in the gut microbiome, potentially influencing the observed reduction in carotenoid-associated skin pigmentation. Our study emphasizes that skin pigmentation is not only impacted by genetic factors but also by the gut microbiome. Meanwhile, the gut microbiome's adaptability can be rapidly shaped and may be driven by specific single-gene variations.
Collapse
Affiliation(s)
- Nusrat Hasan Kanika
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Xin Hou
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Hao Liu
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Yue Dong
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Jun Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Chenghui Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
29
|
Yan R, Zeng X, Shen J, Wu Z, Guo Y, Du Q, Tu M, Pan D. New clues for postbiotics to improve host health: a review from the perspective of function and mechanisms. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6376-6387. [PMID: 38450745 DOI: 10.1002/jsfa.13444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/08/2024]
Abstract
Strain activity and stability severely limit the beneficial effects of probiotics in modulating host health. Postbiotics have emerged as a promising alternative as they can provide similar or even enhanced efficacy to probiotics, even under inactivated conditions. This review introduces the ingredients, preparation, and identification techniques of postbiotics, focusing on the comparison of the advantages and limitations between probiotics and postbiotics based on their mechanisms and applications. Inactivation treatment is the most significant difference between postbiotics and probiotics. We highlight the use of emerging technologies to inactivate probiotics, optimize process conditions to maintain the activity of postbiotics, or scale up their production. Postbiotics have high stability which can overcome unfavorable factors, such as easy inactivation and difficult colonization of probiotics after entering the intestine, and are rapidly activated, allowing continuous and rapid optimization of the intestinal microecological environment. They provide unique mechanisms, and multiple targets act on the gut-organ axis, co-providing new clues for the study of the biological functions of postbiotics. We summarize the mechanisms of action of inactivated lactic acid bacteria, highlighting that the NF-κB and MAPK pathways can be used as immune targeting pathways for postbiotic modulation of host health. Generally, we believe that as the classification, composition, and efficacy mechanism of postbiotics become clearer they will be more widely used in food, medicine, and other fields, greatly enriching the dimensions of food innovation. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ruonan Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Xiaoqun Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Jiamin Shen
- Zhejiang Shenjinji Food Technology Co., LTD, Huzhou, China
| | - Zhen Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Yuxing Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Qiwei Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Maolin Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Insititute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food Science and Engineering, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| |
Collapse
|
30
|
Randeni N, Bordiga M, Xu B. A Comprehensive Review of the Triangular Relationship among Diet-Gut Microbiota-Inflammation. Int J Mol Sci 2024; 25:9366. [PMID: 39273314 PMCID: PMC11394685 DOI: 10.3390/ijms25179366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The human gastrointestinal tract hosts a complex and dynamic community of microorganisms known as the gut microbiota, which play a pivotal role in numerous physiological processes, including digestion, metabolism, and immune function. Recent research has highlighted the significant impact of diet on the gut microbiota composition and functionality, and the consequential effects on host health. Concurrently, there is growing evidence linking the gut microbiota to inflammation, a key factor in many chronic diseases such as inflammatory bowel disease (IBD), obesity, diabetes, and cardiovascular diseases (CVDs). This review explores how dietary components influence the gut microbiota composition, how these microbial changes affect inflammatory pathways, and the therapeutic implications of modulating this axis for chronic inflammatory disease prevention and management. Beneficial dietary patterns, such as the Mediterranean diet (MD) and plant-based diets, promote a diverse and balanced gut microbiota composition, supporting anti-inflammatory pathways. Conversely, the Western diet (WD), high in saturated fats and refined sugars, is associated with dysbiosis and increased inflammation. With all the links between the three variables considered, this review attempts to offer a thorough examination of the triangle formed by inflammation, the gut microbiota, and food.
Collapse
Affiliation(s)
- Nidesha Randeni
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
- Department of Agricultural and Plantation Engineering, Faculty of Engineering Technology, The Open University of Sri Lanka, Nawala, Nugegoda 10250, Sri Lanka
| | - Matteo Bordiga
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
| |
Collapse
|
31
|
Govaert M, Rotsaert C, Vannieuwenhuyse C, Duysburgh C, Medlin S, Marzorati M, Jarrett H. Survival of Probiotic Bacterial Cells in the Upper Gastrointestinal Tract and the Effect of the Surviving Population on the Colonic Microbial Community Activity and Composition. Nutrients 2024; 16:2791. [PMID: 39203927 PMCID: PMC11357584 DOI: 10.3390/nu16162791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Many health-promoting effects have been attributed to the intake of probiotic cells. However, it is important that probiotic cells arrive at the site of their activity in a viable state in order to exert their beneficial effects. Careful selection of the appropriate probiotic formulation is therefore required as mainly the type of probiotic species/strain and the administration strategy may affect survival of the probiotic cells during the upper gastrointestinal (GIT) passage. Therefore, the current study implemented Simulator of the Human Microbial Ecosystem (SHIME®) technology to investigate the efficacy of different commercially available probiotic formulations on the survival and culturability of probiotic bacteria during upper GIT passage. Moreover, Colon-on-a-Plate (CoaP™) technology was applied to assess the effect of the surviving probiotic bacteria on the gut microbial community (activity and composition) of three human donors. Significantly greater survival and culturability rates were reported for the delayed-release capsule formulation (>50%) as compared to the powder, liquid, and standard capsule formulations (<1%) (p < 0.05), indicating that the delayed-release capsule was most efficacious in delivering live bacteria cells. Indeed, administration of the delayed-release capsule probiotic digest resulted in enhanced production of SCFAs and shifted gut microbial community composition towards beneficial bacterial species. These results thus indicate that careful selection of the appropriate probiotic formulation and administration strategy is crucial to deliver probiotic cells in a viable state at the site of their activity (distal ileum and colon).
Collapse
Affiliation(s)
- Marlies Govaert
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.G.); (C.R.); (C.V.)
| | - Chloë Rotsaert
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.G.); (C.R.); (C.V.)
| | | | - Cindy Duysburgh
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.G.); (C.R.); (C.V.)
| | - Sophie Medlin
- Heights, Department for Research and Development, London W1D 2LG, UK; (S.M.); (H.J.)
| | - Massimo Marzorati
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (M.G.); (C.R.); (C.V.)
- Center of Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Harry Jarrett
- Heights, Department for Research and Development, London W1D 2LG, UK; (S.M.); (H.J.)
| |
Collapse
|
32
|
Biswas M, Nurunnabi M, Khatun Z. Understanding Mucosal Physiology and Rationale of Formulation Design for Improved Mucosal Immunity. ACS APPLIED BIO MATERIALS 2024; 7:5037-5056. [PMID: 38787767 DOI: 10.1021/acsabm.4c00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The oral and nasal cavities serve as critical gateways for infectious pathogens, with microorganisms primarily gaining entry through these routes. Our first line of defense against these invaders is the mucosal membrane, a protective barrier that shields the body's internal systems from infection while also contributing to vital functions like air and nutrient intake. One of the key features of this mucosal barrier is its ability to protect the physiological system from pathogens. Additionally, mucosal tolerance plays a crucial role in maintaining homeostasis by regulating the pH and water balance within the body. Recognizing the importance of the mucosal barrier, researchers have developed various mucosal formulations to enhance the immune response. Mucosal vaccines, for example, deliver antigens directly to mucosal tissues, triggering local immune stimulation and ultimately inducing systemic immunity. Studies have shown that lipid-based formulations such as liposomes and virosomes can effectively elicit both local and systemic immune responses. Furthermore, mucoadhesive polymeric particles, with their prolonged delivery to target sites, have demonstrated an enhanced immune response. This Review delves into the critical role of material selection and delivery approaches in optimizing mucosal immunity.
Collapse
Affiliation(s)
- Mila Biswas
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| |
Collapse
|
33
|
Bárcenas-Preciado V, Mata-Haro V. Probiotics in miRNA-Mediated Regulation of Intestinal Immune Homeostasis in Pigs: A Physiological Narrative. Microorganisms 2024; 12:1606. [PMID: 39203448 PMCID: PMC11356641 DOI: 10.3390/microorganisms12081606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
The microbiota plays a crucial role in maintaining the host's intestinal homeostasis, influencing numerous physiological functions. Various factors, including diet, stress, and antibiotic use, can lead to such imbalances. Probiotics have been shown to restore the microbiota, contributing to maintaining this balance. For instance, the weaning stage in piglets is crucial; this transition can cause unfavorable changes that may contribute to the onset of diarrhea. Probiotic supplementation has increased due to its benefits. However, its mechanism of action is still controversial; one involves the regulation of intestinal immunity. When recognized by immune system cells through membrane receptors, probiotics activate intracellular signaling pathways that lead to changes in gene expression, resulting in an anti-inflammatory response. This complex regulatory system involves transcriptional and post-transcriptional mechanisms, including the modulation of various molecules, emphasizing microRNAs. They have emerged as important regulators of innate and adaptive immune responses. Analyzing these mechanisms can enhance our understanding of probiotic-host microbiota interactions, providing insights into their molecular functions. This knowledge can be applied not only in the swine industry, but also in studying microbiota-related disorders. Moreover, these studies serve as animal models, helping to understand better conditions such as inflammatory bowel disease and other related disorders.
Collapse
Affiliation(s)
| | - Verónica Mata-Haro
- Laboratorio de Microbiología e Inmunología, Centro de Investigación en Alimentación y Desarrollo, AC (CIAD) Carretera Gustavo E. Astiazarán 46, Col. La Victoria, Hermosillo 83304, Mexico;
| |
Collapse
|
34
|
Kerstens R, Joyce P. The Gut Microbiome as a Catalyst and Emerging Therapeutic Target for Parkinson's Disease: A Comprehensive Update. Biomedicines 2024; 12:1738. [PMID: 39200203 PMCID: PMC11352163 DOI: 10.3390/biomedicines12081738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's Disease is the second most prevalent neurological disorder globally, and its cause is still largely unknown. Likewise, there is no cure, and existing treatments do little more than subdue symptoms before becoming ineffective. It is increasingly important to understand the factors contributing to Parkinson's Disease aetiology so that new and more effective pharmacotherapies can be established. In recent years, there has been an emergence of research linking gut dysbiosis to Parkinson's Disease via the gut-brain axis. Advancements in microbial profiling have led to characterisation of a Parkinson's-specific microbial signature, where novel treatments that leverage and correct gut dysbiosis are beginning to emerge for the safe and effective treatment of Parkinson's Disease. Preliminary clinical studies investigating microbiome-targeted therapeutics for Parkinson's Disease have revealed promising outcomes, and as such, the aim of this review is to provide a timely and comprehensive update of the most recent advances in this field. Faecal microbiota transplantation has emerged as a novel and potential frontrunner for microbial-based therapies due to their efficacy in alleviating Parkinson's Disease symptomology through modulation of the gut-brain axis. However, more rigorous clinical investigation, along with technological advancements in diagnostic and in vitro testing tools, are critically required to facilitate the widespread clinical translation of microbiome-targeting Parkinson's Disease therapeutics.
Collapse
Affiliation(s)
| | - Paul Joyce
- Centre for Pharmaceutical Innovation (CPI), UniSA Clinical & Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| |
Collapse
|
35
|
Fuentes J, Gregório SF, Fonseca F, Robles-Arozarena R, Martos-Sitcha JA, Moyano FJ. Effect of bile salts on intestinal epithelial function in gilthead seabream (Sparus aurata). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1777-1790. [PMID: 38916718 PMCID: PMC11286703 DOI: 10.1007/s10695-024-01369-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/16/2024] [Indexed: 06/26/2024]
Abstract
In the context of modern aquaculture, the effort to reduce the reliance on fishmeal/marine ingredients in fish diets has led to the exploration of plant-based protein sources as potential substitutes, a dietary shift that disrupts the bile acid profile in fish. Therefore, bile salts are being sought as additives. However, artificially increased intestinal levels of bile acids may significantly impact mucosal function. Therefore, here, we explored the regulatory role in the intestine of gilthead sea bream (Sparus aurata) of (i) chenodeoxycholic acid (CDC), (ii) a mixture formed by two bile acids, 3% cholic acid and 97% deoxycholic acid (MIX), and (iii) a conjugated bile salt sodium taurocholate (TC) in Ussing chambers with the epithelial voltage clamp technique. We tested the bile salts in a 50-500 μg/ml concentration range, and all of them promoted ion absorption. Yet, clear concentration-dependent and more pronounced effects on the ion transport were observed in the posterior intestine. On the other hand, bile salts had no or minor effects on tissue resistance. However, there are indications that the MIX could have adverse effects at high concentrations (500 μg/ml), promoting a threefold increase in tissue permeability measured using FITC-dextran (4 kD) regardless of the intestinal region, thus suggesting an alteration in intestinal permeability at high bile salt concentrations. The findings from our study emphasize the importance of considering intestinal function when contemplating the possible use of a particular bile salt as a dietary supplement. It appears that bile salts, whether acting individually or in combination, play a pivotal role in orchestrating nutrient absorption by influencing the function of epithelial ion transport. However further research is needed to fully grasp the region-dependent nuances of bile salt effects on ion transport and the ultimate consequences for nutrient absorption in the context of fish aquaculture.
Collapse
Affiliation(s)
- J Fuentes
- Consejo Superior de Investigaciones Científicas (ICMAN-CSIC), Instituto de Ciencias Marinas de Andalucía, 11519, Puerto Real, Cádiz, Spain.
| | - S F Gregório
- Centro de Ciências do Mar, Universidade do Algarve, 8005-139, Faro, Portugal
| | - F Fonseca
- ARNET (Aquatic Network, Associated Laboratory), Centre for Marine and Environmental Research (CIMA), University of Algarve, Faro, Portugal
| | | | - J A Martos-Sitcha
- Departamento de Biología Facultad de Ciencias del Mar y Ambientales, Instituto Universitario de Investigación Marina (INMAR), Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, 11519, Puerto Real, Cádiz, Spain
| | - F J Moyano
- Departamento de Biologia y Geologia Facultad de Ciencias, Campus de Excelencia Internacional del Mar (CEI-MAR), Universidad de Almeria, La Cañada de San Urbano, 04120, Almería, Spain.
| |
Collapse
|
36
|
Ge C, Luo X, Lv Y, Wu L, Hu Z, Huang W, Zhan S, Shen X, Hui C, Yu D, Liu B. Essential oils ameliorate the intestinal damages induced by nonylphenol exposure by modulating tryptophan metabolism and activating aryl hydrocarbon receptor via gut microbiota regulation. CHEMOSPHERE 2024; 362:142571. [PMID: 38876325 DOI: 10.1016/j.chemosphere.2024.142571] [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: 02/29/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Nonylphenol (NP) is a ubiquitous endocrine disruptor that persists in the environment and can significantly contribute to serious health hazards, particularly intestinal barrier injury. Plant essential oils (EOs) have recently gained widespread interest due to their potential for improving intestinal health. However, the precise mechanism and protective effects of EOs ameliorating the intestinal damages induced by NP exposure remain unclear. To clarify the potential mechanism and protective impact of EOs against intestinal injury induced by NP, a total of 144 one-day-old male ducks were randomly allocated to four groups: CON (basal diet), EO (basal diet + 200 mg/kg EOs), NP (basal diet + 40 mg/kg NP), and NPEO (basal diet + 200 mg/kg EOs + 40 mg/kg NP). The data revealed that NP exposure significantly damaged intestinal barrier, as evidenced by a reduction in the levels of tight junction gene expression and an increase in intestinal permeability. Additionally, it disturbed gut microbiota, as well as interfered with tryptophan (Trp) metabolism. The NP-induced disorder of Trp metabolism restrained the activation of aryl hydrocarbon receptor (AhR) and resulted in decreased the expression levels of CYP1A1, IL-22, and STAT3 genes, which were alleviated after treatment with EOs. Taken together, NP exposure resulted in impairment of the intestinal barrier function, disruption of gut microbiota, and disturbances in Trp metabolism. Dietary EOs supplementation alleviated the intestinal barrier injury induced by NP through the Trp/AhR/IL-22 signaling pathway.
Collapse
Affiliation(s)
- Chaoyue Ge
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China; Hainan Institute, Zhejiang University, Sanya, 572000, China
| | - Xinyu Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yujie Lv
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China; Hainan Institute, Zhejiang University, Sanya, 572000, China
| | - Lianchi Wu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhaoying Hu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Weichen Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shenao Zhan
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyu Shen
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Cai Hui
- School of Environmental Science and Engineering, Zhejiang Gongshang University, Hangzhou, 310012, China
| | - Dongyou Yu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China; Hainan Institute, Zhejiang University, Sanya, 572000, China.
| | - Bing Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
37
|
Munteanu C, Schwartz B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int J Mol Sci 2024; 25:8250. [PMID: 39125822 PMCID: PMC11311432 DOI: 10.3390/ijms25158250] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The intricate relationship between the gastrointestinal (GI) microbiome and the progression of chronic non-communicable diseases underscores the significance of developing strategies to modulate the GI microbiota for promoting human health. The administration of probiotics and prebiotics represents a good strategy that enhances the population of beneficial bacteria in the intestinal lumen post-consumption, which has a positive impact on human health. In addition, dietary fibers serve as a significant energy source for bacteria inhabiting the cecum and colon. Research articles and reviews sourced from various global databases were systematically analyzed using specific phrases and keywords to investigate these relationships. There is a clear association between dietary fiber intake and improved colon function, gut motility, and reduced colorectal cancer (CRC) risk. Moreover, the state of health is reflected in the reciprocal and bidirectional relationships among food, dietary antioxidants, inflammation, and body composition. They are known for their antioxidant properties and their ability to inhibit angiogenesis, metastasis, and cell proliferation. Additionally, they promote cell survival, modulate immune and inflammatory responses, and inactivate pro-carcinogens. These actions collectively contribute to their role in cancer prevention. In different investigations, antioxidant supplements containing vitamins have been shown to lower the risk of specific cancer types. In contrast, some evidence suggests that taking antioxidant supplements can increase the risk of developing cancer. Ultimately, collaborative efforts among immunologists, clinicians, nutritionists, and dietitians are imperative for designing well-structured nutritional trials to corroborate the clinical efficacy of dietary therapy in managing inflammation and preventing carcinogenesis. This review seeks to explore the interrelationships among dietary antioxidants, dietary fiber, and the gut microbiome, with a particular focus on their potential implications in inflammation and cancer.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
38
|
Balendra V, Rosenfeld R, Amoroso C, Castagnone C, Rossino MG, Garrone O, Ghidini M. Postbiotics as Adjuvant Therapy in Cancer Care. Nutrients 2024; 16:2400. [PMID: 39125280 PMCID: PMC11314502 DOI: 10.3390/nu16152400] [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: 06/04/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Postbiotics are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit to the host. They range from cell wall fragments to metabolites, bacterial lysates, extracellular vesicles, and short-chain fatty acids (SCFAs). Postbiotics may influence carcinogenesis via a variety of mechanisms. They can promote homeostatic immune responses, reduce inflammation, induce selective cytotoxicity against tumor cells, as well as the enabling the control of tumor cell proliferation and enhancing intestinal epithelial barrier function. Therefore, probiotics can serve as an adjunct strategy in anticancer treatment together with chemotherapy and immunotherapy. Up to now, the only relevant postbiotics used as interventions in oncological patients remain vitamin K molecules, with few phase-II and III trials available. In fact, postbiotics' levels are strictly dependent on the gut microbiota's composition, which may vary between individuals and can be altered under different physiological and pathological conditions. Therefore, the lack of consistent clinical evidence supporting postbiotics' efficacy is due to their poor bioavailability, short half-life, and fluctuating levels. Synbiotics, a mixture of prebiotics and probiotics, are expected to have a more homogeneous bioavailability with respect to postbiotics and may have greater potential for future development. In this review, we focus on the role of postbiotics as an adjuvant therapy in cancer treatment.
Collapse
Affiliation(s)
| | - Roberto Rosenfeld
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | | | - Maria Grazia Rossino
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Ornella Garrone
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| |
Collapse
|
39
|
Zhang R, Chen Y, Feng Z, Cai B, Cheng Y, Du Y, Ou S, Chen H, Pan M, Liu H, Pei D, Cao S. Reprogramming human urine cells into intestinal organoids with long-term expansion ability and barrier function. Heliyon 2024; 10:e33736. [PMID: 39040281 PMCID: PMC11261862 DOI: 10.1016/j.heliyon.2024.e33736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Generation of intestinal organoids from human somatic cells by reprogramming would enable intestinal regeneration, disease modeling, and drug screening in a personalized pattern. Here, we report a direct reprogramming protocol for the generation of human urine cells induced intestinal organoids (U-iIOs) under a defined medium. U-iIOs expressed multiple intestinal specific genes and showed resembling gene expression profiles to primary small intestines. U-iIOs can be stably long-term expanded and further differentiated into more mature intestinal lineage cells with high expression of metallothionein and cytochrome P450 (CYP450) genes. These specific molecular features of U-iIOs differ from human pluripotent stem cells derived intestinal organoids (P-iIOs) and intestinal immortalized cell lines. Furthermore, U-iIOs exhibit intestinal barriers indicated by blocking FITC-dextran permeation and uptaking of the specific substrate rhodamine 123. Our study provides a novel platform for patient-specific intestinal organoid generation, which may lead to precision treatment of intestinal diseases and facilitate drug discovery.
Collapse
Affiliation(s)
- Ruifang Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Yating Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Ziyu Feng
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Baomei Cai
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Yiyi Cheng
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunjing Du
- School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Sihua Ou
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Huan Chen
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Mengjie Pan
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - He Liu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shangtao Cao
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| |
Collapse
|
40
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [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: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
41
|
Ávila-Arres IE, Rodríguez Hernández E, Gómez Rosales S, Reis de Souza TC, Mariscal-Landín G. Proteomic Identification and Quantification of Basal Endogenous Proteins in the Ileal Digesta of Growing Pigs. Animals (Basel) 2024; 14:2000. [PMID: 38998112 PMCID: PMC11240675 DOI: 10.3390/ani14132000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
The accurate estimation of basal endogenous losses (BEL) of amino acids at the ileum is indispensable to improve nutrient utilization efficiency. This study used a quantitative proteomic approach to identify variations in BEL in the ileal digesta of growing pigs fed a nitrogen-free diet (NFD) or a casein diet (CAS). Eight barrow pigs (39.8 ± 6.3 kg initial body weight (BW)) were randomly assigned to a 2 × 2 crossover design. A total of 348 proteins were identified and quantified in both treatments, of which 101 showed a significant differential abundance between the treatments (p < 0.05). Functional and pathway enrichment analyses revealed that the endogenous proteins were associated with intestinal metabolic function. Furthermore, differentially abundant proteins (DAPs) in the digesta of pigs fed the NFD enriched terms and pathways that suggest intestinal inflammation, the activation of innate antimicrobial host defense, an increase in cellular autophagy and epithelial turnover, and reduced synthesis of pancreatic and intestinal secretions. These findings suggest that casein diets may provide a more accurate estimation of BEL because they promote normal gastrointestinal secretions. Overall, proteomic and bioinformatic analyses provided valuable insights into the composition of endogenous proteins in the ileal digesta and their relationship with the functions, processes, and pathways modified by diet composition.
Collapse
Affiliation(s)
- Iris Elisa Ávila-Arres
- Posgrado en Ciencias de la Producción y de la Salud Animal, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Elba Rodríguez Hernández
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Km 1 Carretera a Colón, Querétaro 76280, Mexico; (E.R.H.); (S.G.R.)
| | - Sergio Gómez Rosales
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Km 1 Carretera a Colón, Querétaro 76280, Mexico; (E.R.H.); (S.G.R.)
| | - Tércia Cesária Reis de Souza
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Av. de las Ciencias s/n, Querétaro 76230, Mexico;
| | - Gerardo Mariscal-Landín
- Centro Nacional de Investigación Disciplinaria en Fisiología y Mejoramiento Animal, INIFAP, Km 1 Carretera a Colón, Querétaro 76280, Mexico; (E.R.H.); (S.G.R.)
| |
Collapse
|
42
|
Jia X, Huang Y, Liu G, Li Z, Tan Q, Zhong S. The Use of Polysaccharide AOP30 from the Rhizome of Alpinia officinarum Hance to Alleviate Lipopolysaccharide-Induced Intestinal Epithelial Barrier Dysfunction and Inflammation via the TLR4/NfκB Signaling Pathway in Caco-2 Cell Monolayers. Nutrients 2024; 16:2151. [PMID: 38999898 PMCID: PMC11243348 DOI: 10.3390/nu16132151] [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: 04/21/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Alpinia officinarum Hance is rich in carbohydrates and is flavored by natives. The polysaccharide fraction 30 is purified from the rhizome of A. officinarum Hance (AOP30) and shows excellent immunoregulatory ability when administered to regulate immunity. However, the effect of AOP30 on the intestinal epithelial barrier is not well understood. Therefore, the aim of this study is to investigate the protective effect of AOP30 on the intestinal epithelial barrier using a lipopolysaccharide (LPS)-induced intestinal epithelial barrier dysfunction model and further explore its underlying mechanisms. Cytotoxicity, transepithelial electrical resistance (TEER) values, and Fluorescein isothiocyanate (FITC)-dextran flux are measured. Simultaneously, the protein and mRNA levels of tight junction (TJ) proteins, including zonula occludens-1 (ZO-1), Occludin, and Claudin-1, are determined using Western blotting and reverse-transcription quantitative polymerase chain reaction methods, respectively. The results indicate that AOP30 restores the LPS-induced decrease in the TEER value and cell viability. Furthermore, it increases the mRNA and protein expression of ZO-1, Occludin, and Claudin-1. Notably, ZO-1 is the primary tight junction protein altered in response to LPS-induced intestinal epithelial dysfunction. Additionally, AOP30 downregulates the production of TNFα via the Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Collectively, the findings of this study indicate that AOP30 can be developed as a functional food ingredient or natural therapeutic agent for addressing intestinal epithelial barrier dysfunction. It sheds light on the role of AOP30 in improving intestinal epithelial function.
Collapse
Affiliation(s)
- Xuejing Jia
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yun Huang
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guanghuo Liu
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zipeng Li
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qiwei Tan
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
43
|
Vasilev GV, Miteva D, Gulinac M, Chervenkov L, Kitanova M, Velikova T. Exploring Gut–Brain Interaction Disorders: Mechanisms and Translational Therapies Crossing Neurology to Gastroenterology. GASTROENTEROLOGY INSIGHTS 2024; 15:555-573. [DOI: 10.3390/gastroent15030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
The bidirectional communication network between the gut and the brain, known as the gut–brain axis, plays a crucial role in health and disease. This review explores the mechanisms underlying gut–brain interaction disorders and highlights translational therapies bridging neurology and gastroenterology. Mechanisms encompass anatomical, endocrine, humoral, metabolic, and immune pathways, with the gut microbiota exerting profound influence. Clinical evidence links gut microbiota fluctuations to mood disorders, GI disruptions, and neurodevelopmental conditions, emphasizing the microbiome’s pivotal role in shaping brain–gut interactions. Pharmacological therapies such as amitriptyline and selective serotonin reuptake inhibitors modulate neurotransmitter activity, offering relief in functional gastrointestinal disorders like irritable bowel syndrome (IBS). Non-pharmacological interventions like cognitive–behavioral therapy and hypnotherapy address maladaptive thoughts and induce relaxation, alleviating gastrointestinal symptoms exacerbated by stress. Emerging therapies include gut microbiota modulation, dietary interventions, vagus nerve stimulation, and intestinal barrier modulation, offering novel approaches to manage neurological disorders via the gastrointestinal tract. Understanding and harnessing the gut–brain axis holds promise for personalized therapeutic strategies in neurogastroenterology.
Collapse
Affiliation(s)
- Georgi V. Vasilev
- Clinic of Neurology and Department of Emergency Medicine, UMHAT “Sv. Georgi”, Blvd. Peshtersko Shose 66, 4000 Plovdiv, Bulgaria
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Dimitrina Miteva
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Lyubomir Chervenkov
- Department of Diagnostic Imaging, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Research Complex for Translational Neuroscience, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4002 Plovdiv, Bulgaria
| | - Meglena Kitanova
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
| |
Collapse
|
44
|
Cheng Y, Xiao X, Fu J, Zong X, Lu Z, Wang Y. Escherichia coli K88 activates NLRP3 inflammasome-mediated pyroptosis in vitro and in vivo. Biochem Biophys Rep 2024; 38:101665. [PMID: 38419757 PMCID: PMC10900769 DOI: 10.1016/j.bbrep.2024.101665] [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: 04/17/2023] [Revised: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Pyroptosis induced by lipopolysaccharide (LPS) has an obvious impact on intestinal inflammation and immune regulation. Enterotoxigenic Escherichia coli (ETEC) K88 has been proved to induce inflammatory responses in several models, but whether E. coli K88 participates in the same process of pyroptotic cell death as LPS remains to be identified. We conducted a pilot experiment to confirm that E. coli K88, instead of Escherichia coli O157 and Salmonella typhimurium, promotes the secretion of interleukin-1 beta (IL-1β) and interleukin-18 (IL-18) in macrophages. Further experiments were carried out to dissect the molecular mechanism both in vitro and in vivo. The Enzyme-Linked Immunosorbent Assay (ELISA) results suggested that E. coli K88 treatment increased the expression of pro-inflammatory cytokines IL-18 and IL-1β in both C57BL/6 mice and the supernatant of J774A.1 cells. Intestinal morphology observations revealed that E. coli K88 treatment mainly induced inflammation in the colon. Real-time PCR and Western blot analysis showed that the mRNA and protein expressions of pyroptosis-related factors, such as NLRP3, ASC, and Caspase1, were significantly upregulated by E. coli K88 treatment. The RNA-seq results confirmed that the effect was associated with the activation of NLRP3, ASC, Caspase1, GSDMD, IL-18, and IL-1β, and might also be related to inflammatory bowel disease and the tumor necrosis factor pathway. The pyroptosis-activated effect of E. coli K88 was significantly blocked by NLRP3 siRNA. Our data suggested that E. coli K88 caused inflammation by triggering pyroptosis, which provides a theoretical basis for the prevention and treatment of ETEC in intestinal infection.
Collapse
Affiliation(s)
- Yuanzhi Cheng
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Xiao Xiao
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang A&F University, Hangzhou, 311300, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Zeqing Lu
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, 310058, China
| |
Collapse
|
45
|
Marino M, Rendine M, Venturi S, Porrini M, Gardana C, Klimis-Zacas D, Riso P, Del Bo' C. Red raspberry ( Rubus idaeus) preserves intestinal barrier integrity and reduces oxidative stress in Caco-2 cells exposed to a proinflammatory stimulus. Food Funct 2024; 15:6943-6954. [PMID: 38855989 DOI: 10.1039/d4fo01050g] [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: 06/11/2024]
Abstract
Growing evidence showed the capacity of (poly)phenols to exert a protective role on intestinal health. Nevertheless, the existing findings are still heterogeneous and the underlying mechanisms remain unclear. This study investigated the potential benefits of a red raspberry (Rubus idaeus) powder on the integrity of the intestinal barrier, focusing on its ability to mitigate the effects of tumor necrosis factor-α (TNF-α)-induced intestinal permeability. Human colorectal adenocarcinoma cells (i.e., Caco-2 cells) were used as a model to assess the impact of red raspberry on intestinal permeability, tight junction expression, and oxidative stress. The Caco-2 cells were differentiated into polarized monolayers and treated with interferon-γ (IFN-γ) (10 ng mL-1) for 24 hours, followed by exposure to TNF-α (10 ng mL-1) in the presence or absence of red raspberry extract (1-5 mg mL-1). The integrity of the intestinal monolayer was evaluated using transepithelial electrical resistance (TEER) and fluorescein isothiocyanate-dextran (FITC-D) efflux assay. Markers of intestinal permeability (claudin-1, occludin, and zonula occludens-1 (ZO-1)) and oxidative stress (8-hydroxy-2-deoxyguanosine (8-OHdG) and protein carbonyl) were assessed using ELISA kits. Treatment with red raspberry resulted in a significant counteraction of TEER value loss (41%; p < 0.01) and a notable reduction in the efflux of FITC-D (-2.5 times; p < 0.01). Additionally, red raspberry attenuated the levels of 8-OHdG (-48.8%; p < 0.01), mitigating the detrimental effects induced by TNF-α. Moreover, red raspberry positively influenced the expression of the integral membrane protein claudin-1 (+18%; p < 0.01), an essential component of tight junctions. These findings contribute to the growing understanding of the beneficial effects of red raspberry in the context of the intestinal barrier. The effect of red raspberry against TNF-α-induced intestinal permeability observed in our in vitro model suggests, for the first time, its potential as a dietary strategy to promote gastrointestinal health.
Collapse
Affiliation(s)
- Mirko Marino
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | - Marco Rendine
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | - Samuele Venturi
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | - Marisa Porrini
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | - Claudio Gardana
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | | | - Patrizia Riso
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| | - Cristian Del Bo'
- Università degli Studi di Milano, DeFENS - Department of Food, Environmental and Nutritional Sciences, Via Celoria 2, 20133 Milano, Italy.
| |
Collapse
|
46
|
Noori E, Hashemi N, Rezaee D, Maleki R, Shams F, Kazemi B, Bandepour M, Rahimi F. Potential therapeutic options for celiac Disease: An update on Current evidence from Gluten-Free diet to cell therapy. Int Immunopharmacol 2024; 133:112020. [PMID: 38608449 DOI: 10.1016/j.intimp.2024.112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
Celiac disease (CD) is a chronic autoimmune enteropathy and multifactorial disease caused by inappropriate immune responses to gluten in the small intestine. Weight loss, anemia, osteoporosis, arthritis, and hepatitis are among the extraintestinal manifestations of active CD. Currently, a strict lifelong gluten-free diet (GFD) is the only safe, effective, and available treatment. Despite the social burden, high expenses, and challenges of following a GFD, 2 to 5 percent of patients do not demonstrate clinical or pathophysiological improvement. Therefore, we need novel and alternative therapeutic approaches for patients. Innovative approaches encompass a broad spectrum of strategies, including enzymatic degradation of gluten, inhibition of intestinal permeability, modulation of the immune response, inhibition of the transglutaminase 2 (TG2) enzyme, blocking antigen presentation by HLA-DQ2/8, and induction of tolerance. Hence, this review is focused on comprehensive therapeutic strategies ranging from dietary approaches to novel methods such as antigen-based immunotherapy, cell and gene therapy, and the usage of nanoparticles for CD treatment.
Collapse
Affiliation(s)
- Effat Noori
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran; Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Maleki
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Forough Shams
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandepour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fardin Rahimi
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
47
|
Jirillo E, Topi S, Charitos IA, Santacroce L, Gaxhja E, Colella M. Gut Microbiota and Immune System in Necrotizing Enterocolitis and Related Sepsis. GASTROINTESTINAL DISORDERS 2024; 6:431-445. [DOI: 10.3390/gidisord6020029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
A severe condition of sepsis can be a complication of necrotizing enterocolitis (NEC), which can occur in premature infants and becomes a medical challenge in the neonatal intensive care unit (NICU). It is a multifactorial intestinal disease (can affect both the small and large intestine) that can lead to ischemia of the intestinal tissues that evolves into acute organ necrosis. One of these factors is that different types of nutrition can influence the onset or the progression of the disease. Cow-milk-based infant formulas have been shown to cause it in premature infants more frequently than human milk. Recently, nutrition has been shown to be beneficial after surgery. Several issues still under study, such as the pathogenesis and the insufficient and often difficult therapeutic approach, as well as the lack of a common and effective prevention strategy, make this disease an enigma in daily clinical practice. Recent studies outlined the emerging role of the host immune system and resident gut microbiota, showing their close connection in NEC pathophysiology. In its initial stages, broad-spectrum antibiotics, bowel rest, and breastfeeding are currently used, as well as probiotics to help the development of the intestinal microbiota and its eubiosis. This paper aims to present the current knowledge and potential fields of research in NEC pathophysiology and therapeutic assessment.
Collapse
Affiliation(s)
- Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Skender Topi
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
| | - Elona Gaxhja
- Department of Clinical Disciplines, School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari, 70124 Bari, Italy
- Doctoral School, eCampus University, 22060 Novedrate, Italy
| |
Collapse
|
48
|
Hearn-Yeates F, Horne AW, O’Mahony SM, Saunders PTK. The impact of the microbiota-gut-brain axis on endometriosis-associated symptoms: mechanisms and opportunities for personalised management strategies. REPRODUCTION AND FERTILITY 2024; 5:RAF-23-0085. [PMID: 38739749 PMCID: PMC11227073 DOI: 10.1530/raf-23-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/13/2024] [Indexed: 05/16/2024] Open
Abstract
Endometriosis is a chronic inflammatory condition affecting one in 10 women and those assigned female at birth, defined by the presence of endometrial-like tissue outside the uterus. It is commonly associated with pain, infertility, and mood disorders, and often comorbid with other chronic pain conditions, such as irritable bowel syndrome. Recent research has identified a key role for the microbiota-gut-brain axis in health and a range of inflammatory and neurological disorders, prompting an exploration of its potential mechanistic role in endometriosis. Increased awareness of the impact of the gut microbiota within the patient community, combined with the often-detrimental side effects of current therapies, has motivated many to utilise self-management strategies, such as dietary modification and supplements, despite a lack of robust clinical evidence. Current research has characterised the gut microbiota in endometriosis patients and animal models. However, small cohorts and differing methodology has resulted in little consensus in the data. In this narrative review, we summarise research studies that have investigated the role of gut microbiota and their metabolic products in the development and progression of endometriosis lesions, before summarising insights from research into co-morbid conditions and discussing the reported impact of self-management strategies on symptoms of endometriosis. Finally, we suggest ways in which this promising field of research could be expanded to explore the role of specific bacteria, improve access to 'microbial' phenotyping, and to develop personalised patient advice for reduction of symptoms such as chronic pain and bloating.
Collapse
Affiliation(s)
- Francesca Hearn-Yeates
- EXPPECT Edinburgh and Centre for Reproductive Health, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| | - Andrew W Horne
- EXPPECT Edinburgh and Centre for Reproductive Health, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| | - Siobhain M O’Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Philippa T K Saunders
- EXPPECT Edinburgh and Centre for Reproductive Health, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| |
Collapse
|
49
|
Gao Y, Liu L, Cui Y, Zhang J, Wu X. The causality of gut microbiota on onset and progression of sepsis: a bi-directional Mendelian randomization analysis. Front Immunol 2024; 15:1266579. [PMID: 38698853 PMCID: PMC11063379 DOI: 10.3389/fimmu.2024.1266579] [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: 07/25/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Background Several observational studies have proposed a potential link between gut microbiota and the onset and progression of sepsis. Nevertheless, the causality of gut microbiota and sepsis remains debatable and warrants more comprehensive exploration. Methods We conducted a two-sample Mendelian randomization (MR) analysis to test the causality between gut microbiota and the onset and progression of sepsis. The genome-wide association study (GWAS) summary statistics for 196 bacterial traits were extracted from the MiBioGen consortium, whereas the GWAS summary statistics for sepsis and sepsis-related outcomes came from the UK Biobank. The inverse-variance weighted (IVW) approach was the primary method used to examine the causal association. To complement the IVW method, we utilized four additional MR methods. We performed a series of sensitivity analyses to examine the robustness of the causal estimates. Results We assessed the causality of 196 bacterial traits on sepsis and sepsis-related outcomes. Genus Coprococcus2 [odds ratio (OR) 0.81, 95% confidence interval (CI) (0.69-0.94), p = 0.007] and genus Dialister (OR 0.85, 95% CI 0.74-0.97, p = 0.016) had a protective effect on sepsis, whereas genus Ruminococcaceae UCG011 (OR 1.10, 95% CI 1.01-1.20, p = 0.024) increased the risk of sepsis. When it came to sepsis requiring critical care, genus Anaerostipes (OR 0.49, 95% CI 0.31-0.76, p = 0.002), genus Coprococcus1 (OR 0.65, 95% CI 0.43-1.00, p = 0.049), and genus Lachnospiraceae UCG004 (OR 0.51, 95% CI 0.34-0.77, p = 0.001) emerged as protective factors. Concerning 28-day mortality of sepsis, genus Coprococcus1 (OR 0.67, 95% CI 0.48-0.94, p = 0.020), genus Coprococcus2 (OR 0.48, 95% CI 0.27-0.86, p = 0.013), genus Lachnospiraceae FCS020 (OR 0.70, 95% CI 0.52-0.95, p = 0.023), and genus Victivallis (OR 0.82, 95% CI 0.68-0.99, p = 0.042) presented a protective effect, whereas genus Ruminococcus torques group (OR 1.53, 95% CI 1.00-2.35, p = 0.049), genus Sellimonas (OR 1.25, 95% CI 1.04-1.50, p = 0.019), and genus Terrisporobacter (OR 1.43, 95% CI 1.02-2.02, p = 0.040) presented a harmful effect. Furthermore, genus Coprococcus1 (OR 0.42, 95% CI 0.19-0.92, p = 0.031), genus Coprococcus2 (OR 0.34, 95% CI 0.14-0.83, p = 0.018), and genus Ruminiclostridium6 (OR 0.43, 95% CI 0.22-0.83, p = 0.012) were associated with a lower 28-day mortality of sepsis requiring critical care. Conclusion This MR analysis unveiled a causality between the 21 bacterial traits and sepsis and sepsis-related outcomes. Our findings may help the development of novel microbiota-based therapeutics to decrease the morbidity and mortality of sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Xiuying Wu
- Department of Anesthesia, ShengJing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
50
|
Tian C, Wang L, Liu M, Liu J, Qiu M, Chen Y. Isolation and Identification of Chicken-Derived Lactic Acid Bacteria: In Vitro Probiotic Properties and Antagonistic Effects against Salmonella pullorum, Staphylococcus aureus, and Escherichia coli. Microorganisms 2024; 12:795. [PMID: 38674739 PMCID: PMC11052321 DOI: 10.3390/microorganisms12040795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The development of probiotics capable of quickly colonizing the intestines of animals is important in promoting the healthy growth of livestock. The aim of this study was to screen lactic acid bacteria (LAB) from the intestinal microbiota of chickens with potential applications, and to evaluate their probiotic properties and antagonistic abilities against Salmonella pullorum, Staphylococcus aureus, and Escherichia coli. The results showed that a total of 79 strains with the characteristics of LAB were isolated from the chicken cecum microbiota, of which 7 strains exhibited strong inhibitory activity against S. pullorum, S. aureus, and E. coli. Performing 16s rDNA sequencing revealed that these seven strains were Lactiplantibacillus pentosus (n = 1), Lactiplantibacillus plantarum (n = 3), Lactiplantibacillus paraplantarum (n = 1), Lactiplantibacillus argentoratensis (n = 1), and Lactiplantibacillus fabifermentans (n = 1). Among them, L. pentosus R26 and L. plantarum R32 exhibited superior antibacterial activity. These two strains demonstrated high lactic acid production ability, with survival rates of 86.29% and 87.99% after 3 h of treatment at pH 1.5, 86.66% and 85.52% after 3 h of treatment with 0.5% bile salts, 90.03% and 88.16% after 2 h of treatment with simulated gastric fluid, and 98.92% and 98.22% after 2 h of treatment with simulated intestinal fluid, respectively. Co-cultivation with L. pentosus R26 for 24 h resulted in 50% of the pathogens being antagonized, while almost complete inhibition was observed following 72 h of co-cultivation. In conclusion, L. pentosus R26 and L. plantarum R32 exhibited high antibacterial activity and acid production capability, while also demonstrating satisfactory tolerance to low pH values and high concentrations of bile salts and digestive fluid. The probiotic characteristics and stress resistance of L. pentosus R26 were slightly superior to those of L. plantarum R32, indicating its potential for development as a probiotic.
Collapse
Affiliation(s)
| | | | | | | | | | - Yong Chen
- Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk, College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (C.T.); (L.W.); (M.L.); (J.L.); (M.Q.)
| |
Collapse
|