1
|
Ghosh S, Chorghade R, Diehl RC, Dodge GJ, Bae S, Dugan AE, Halim M, Wuo MG, Bartlett H, Herndon L, Kiessling LL, Imperiali B. Tools for investigating host-microbe crosstalk using glycan analysis probes inspired by human lectins. Glycobiology 2025; 35:cwaf031. [PMID: 40421735 PMCID: PMC12167852 DOI: 10.1093/glycob/cwaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Human lectins are critical carbohydrate-binding proteins that recognize diverse glycoconjugates from microorganisms and can play a key role in host-microbe interactions. Despite their importance in immune recognition and microbe binding, the specific glycan ligands and functions of many human lectins remain poorly understood. Using previous proof-of-concept studies on selected lectins as the foundation for this work, we present ten additional glycan analysis probes (GAPs) from a diverse set of human soluble lectins, offering robust tools to investigate glycan-mediated interactions. We describe a protein engineering platform that enables scalable production of GAPs that maintain native-like conformations and oligomerization states, equipped with functional reporter tags for targeted glycan profiling. We demonstrate that the soluble GAP reagents can be used in various applications, including glycan array analysis, mucin-binding assays, tissue staining, and microbe binding in complex populations. These capabilities make GAPs valuable for dissecting interactions relevant to understanding host responses to microbes. The tools can also be used to probe differential microbial and mammalian glycan interactions, which are crucial for understanding the interactions of lectins in a physiological environment where both glycan types exist. GAPs have potential as diagnostic and prognostic tools for detecting glycan alterations in chronic diseases, microbial dysbiosis, and immune-related conditions.
Collapse
Affiliation(s)
- Soumi Ghosh
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Rajeev Chorghade
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Roger C Diehl
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Greg J Dodge
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Sunhee Bae
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Amanda E Dugan
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Melanie Halim
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Michael G Wuo
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Helen Bartlett
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Liam Herndon
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Barbara Imperiali
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| |
Collapse
|
2
|
Abraham TJ, Bora M, Bardhan A, Sen A, Das R, Nadella RK, Patil PK. In-feed oxolinic acid induces oxidative stress and histopathological alterations in Nile tilapia Oreochromis niloticus. Toxicol Rep 2025; 14:102020. [PMID: 40242397 PMCID: PMC12002751 DOI: 10.1016/j.toxrep.2025.102020] [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: 12/01/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
The aquaculture industry urgently requires effective bacterial disease management strategies, necessitating better regulation of antibiotic application. This study investigated the effects of oral oxolinic acid (OA) administration on Oreochromis niloticus at the recommended dose of 12 mg (1 ×) and overdose of 36 mg (3 ×)/kg biomass/day for 7 consecutive days in terms of growth, oxidative stress, residue accretion and histopathology relative to the control. The 1 × and 3 × groups experienced dose-dependent mortalities (3.33-8.33 %). The OA residues peaked in the liver and kidney tissues with dosing and declined upon discontinuation. The residues persisted in the kidney even on day 35 post-dosing. Elevated malondialdehyde and total nitric oxide levels signified oxidative stress and correlated with the tissue level changes in various organs. Histologically, glycogen-type vacuolation and cellular hypertrophy were observed in the liver. The kidney had hydropic swelling, renal epithelium degradation, nephrocalcinosis, vacuolation, and necrosis. Splenic alterations were confined to necrosis and a slight increase in sinusoidal space. Intestinal tissues exhibited a depletion of absorptive vacuoles, epithelial layer degradation, mucinous degeneration, and necrosis. Gills displayed epithelial hyperplasia, thickening of secondary lamellae, and erosion. Nevertheless, the cohort administered the recommended dose exhibited recovery with OA discontinuation. However, none of the assessed parameters normalized in the overdosed group even after 35 days of dose suspension. The results indicated that O. niloticus can safely adapt to and tolerate the toxic effects of OA. As the recommended dose of OA elicited reversible bioresponses effectively in tilapia, it can be utilized in aquaculture with due caution following regulations.
Collapse
Affiliation(s)
- Thangapalam Jawahar Abraham
- Department of Aquatic Animal Health, Faculty of Fishery Sciences, West Bengal University of Animal and Fishery Sciences, Chakgaria, Kolkata, West Bengal 700094, India
| | - Masud Bora
- Department of Aquatic Animal Health, Faculty of Fishery Sciences, West Bengal University of Animal and Fishery Sciences, Chakgaria, Kolkata, West Bengal 700094, India
| | - Avishek Bardhan
- Department of Aquatic Animal Health, Faculty of Fishery Sciences, West Bengal University of Animal and Fishery Sciences, Chakgaria, Kolkata, West Bengal 700094, India
| | - Arya Sen
- Department of Aquatic Animal Health, Faculty of Fishery Sciences, West Bengal University of Animal and Fishery Sciences, Chakgaria, Kolkata, West Bengal 700094, India
| | - Ratnapriya Das
- Department of Aquatic Animal Health, Faculty of Fishery Sciences, West Bengal University of Animal and Fishery Sciences, Chakgaria, Kolkata, West Bengal 700094, India
| | - Ranjit Kumar Nadella
- Fish Processing Division, ICAR-Central Institute of Fisheries Technology, Willington Island, Cochin, Kerala 682029, India
| | - Prasanna Kumar Patil
- Aquatic Animal Health and Environment Division, ICAR-Central Institute of Brackishwater Aquaculture, Raja Annamalai Puram, Chennai, Tamil Nadu 600028, India
| |
Collapse
|
3
|
Wang T, Bao MY, Xiao GX, Wang Z, Zhou NN, Wei H, Qiao F, Du ZY, Zhang ML. The defatted black soldier fly meal (Hermetia illucens) improved the pathogen resistance and gut health of Nile Tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110242. [PMID: 40024586 DOI: 10.1016/j.fsi.2025.110242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
As a novel protein source, the black soldier fly (Hermetia illucens) possesses the potential to enhance fish health due to its high protein content, essential amino acids, and bioactive compounds, but the mechanisms by which defatted black soldier fly meal affects fish health remain unclear. This study aimed to evaluate the effects of partially replacing soybean meal (SM) with defatted black soldier fly larvae meal on the pathogen resistance of juvenile Nile tilapia (Oreochromis niloticus) and to explore the underlying mechanisms. Defatted black soldier fly (Hermetia illucens) meal was used to replace 0 % (CON), 15 % (H15), 30 % (H30), 45 % (H45), and 60 % (H60) of soybean meal in the diets, which were fed to Nile tilapia for 8 weeks. At the second week of the experiment, an unexpected infection was observed in the Nile tilapia, with subsequent isolation and identification confirming the pathogen as Aeromonas veronii. The results showed that defatted black soldier fly meal improved the survival rate and reduced the intestinal inflammation of Nile tilapia in H60 group compared to CON group (P < 0.05). Periodic acid-Schiff (PAS) staining of the gut indicated that a 60 % replacement of soybean meal with defatted black soldier fly meal significantly increased the number of goblet cells in the intestine (P < 0.05) and upregulated the expression level of mucin 2 (muc2) (P < 0.05). Throughout the experiment period, the survival rate in the defatted black soldier fly meal groups was higher than that in the soybean meal group, with the H60 group exhibiting the most significant improvement. In addition, defatted black soldier fly meal exhibited a promotion effect on goblet cell numbers, mucin production and mucin secretion with a dose dependent manner (P < 0.05). To identify the possible mechanism by which defatted black soldier fly meal enhanced goblet cell numbers, gut microbiome and metabolomics were conducted. The results showed that the defatted black soldier fly meal altered the composition of intestinal microbiota and increased the content of L-tyrosine in the H60 group. The effects of L-tyrosine were further identified in LS174T cells, and the results showed that L-tyrosine upregulated the expression levels of muc2 and SAM pointed domain-containing Ets transcription factor (spdef) (P < 0.05). In conclusion, the defatted black soldier fly meal improves gut health and enhances pathogen resistance, ultimately leading to the increased survival rates of Nile tilapia when exposed to A. veronii.
Collapse
Affiliation(s)
- Tong Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ming-Yang Bao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guan-Xiu Xiao
- Key Laboratory of Microbial Resources Protection, Development and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, China
| | - Zhe Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Nan-Nan Zhou
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Hua Wei
- Ningbo College of Health Sciences, Zhejiang, 315100, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mei-Ling Zhang
- Key Laboratory of Microbial Resources Protection, Development and Utilization, College of Biological Sciences and Technology, Yili Normal University, Yining, 835000, China; Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
4
|
Wang R, Ma F, Yin D, Wang H, Wei X. Intestinal Microbes, Metabolites, and Hormones in Alcohol-Associated Liver Disease. Semin Liver Dis 2025. [PMID: 40334703 DOI: 10.1055/a-2601-9480] [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: 05/09/2025]
Abstract
Alcohol-associated liver disease (ALD)-encompassing conditions including steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma-refers to hepatic damage arising from excessive or hazardous alcohol consumption, and is now recognized as a significant global health burden. Although the mechanisms underlying ALD remain incompletely understood, several pathways have been substantiated over the last five decades, notably the involvement of intestinal microorganisms and the involvement of the gut-liver axis in alcohol metabolism and ALD pathogenesis. Ethanol intake disrupts the intestinal microbial balance and compromises the gut barrier, resulting in increased permeability to microbial products. The subsequent translocation of microbial metabolites and other antigenic substances to the liver activates hepatic immune responses, thereby contributing to liver injury. In addition, gastrointestinal hormones are also implicated in ALD progression through various mechanisms. Although no therapies for ALD have been approved by the Food and Drug Administration, various therapeutic strategies targeting the intestinal microbiota and gut barrier have been identified. In conclusion, this review discusses the role of the gut-liver axis in alcohol metabolism and ALD pathogenesis and explores the emerging therapeutic strategies.
Collapse
Affiliation(s)
- Ruimeng Wang
- Second Clinical Medical College, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Dou Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaohui Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Wang H, Hou L, Chen X, Gui L, Jiang W, Tang W. Clostridium butyricum protects the ileal barrier in mice by regulating the farnesoid X receptor signaling pathway. Int J Biochem Cell Biol 2025; 185:106798. [PMID: 40398713 DOI: 10.1016/j.biocel.2025.106798] [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: 11/01/2024] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND The intestinal barrier has an important role in maintaining homeostasis. The aim of this study was to determine the protective effect of Clostridium butyricum (CBM) on small intestinal barrier damage in mice and the role of farnesoid X receptor (FXR) in regulating the intestinal barrier by C. butyricum. METHODS AND RESULTS A model of small intestinal injury induced by dextran sulfate sodium (DSS) was constructed to detect repair of intestinal barrier damage after feeding with C. butyricum. In the DSS model group, expression of the tight junction protein (TJP) was significantly decreased and expression of inflammatory factors was significantly increased. TJP expression was significantly increased and inflammatory factor expression was significantly decreased after C. butyricum feeding, which indicated that intestinal barrier function was repaired. In addition, inhibition of FXR expression as well as the downstream signaling pathways were demonstrated in the DSS model group. FXR and its downstream signaling pathways were significantly upregulated after feeding with C. butyricum. Then, intestinal barrier function was evaluated by constructing an intestinal-specific FXR knockout (KO) DSS model in mice. Suppression of TJP and upregulated expression of inflammatory factors were detected in the KO DSS group but there was no significant difference in the expression of TJP and inflammatory factors after C. butyricum feeding. Furthermore, there was no significant difference in FXR downstream signaling pathway expression after C. butyricum feeding compared to the KO DSS group. C. butyricum supernatants (CSs) upregulated the FXR signaling pathways in vitro. CSs did not activate the FXR signaling pathway when FXR was suppressed. CONCLUSIONS C. butyricum supplementation effectively ameliorated DSS-induced intestinal barrier disruption. C. butyricum may have a protective effect on the small intestine through the FXR signaling pathway.
Collapse
Affiliation(s)
- Hanfei Wang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Li Hou
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xintong Chen
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Linling Gui
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Jiang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Chang S, Song M, Lee J, Kim S, Song D, Jeon K, Kim H, Yang J, Kim H, Cho J. Stimbiotics help improve intestinal immunity and positively modulate the gut microbiome in broilers with necrotic enteritis. Poult Sci 2025; 104:105315. [PMID: 40466265 PMCID: PMC12167796 DOI: 10.1016/j.psj.2025.105315] [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: 02/16/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 06/19/2025] Open
Abstract
This experiment was conducted to investigate the effect of stimbiotic (STB) in broilers with necrotic enteritis (NE) on nutrient digestibility and gut health. A total of 200 one-day-old Arbor Acres (initial body weight of 44.03 ± 0.28 g) were used in this experiment for 28 days. All broilers were randomly allocated into four treatments, and each experimental group had 10 replicate cages with five broilers per cage. The experiment was conducted in a 2 × 2 factorial designs consisting of two levels of challenge (challenge and non-challenge) and two levels of STB (0 and 0.05 %). All broilers in challenged groups were orally challenged by overdosing with coccidia vaccines (× 10 recommended doses; Livacox® Q). The NE challenge significantly decreased (P < 0.05) nutrient digestibility, interferon-γ, heterophil levels in blood, and villus height:crypt depth (VH:CD) compared to the non-challenge group. Also, the NE challenge significantly lower (P < 0.05) ZO-1 and higher MUC2 gene expression than the non-challenge group. Supplementation of 0.05 % STB with NE challenge significantly increased (P < 0.05) gross energy digestibility and decreased (P < 0.05) the number of oocysts per gram of feces compared to the NE-challenged group. Supplementation of 0.05 % STB significantly increased (P < 0.05) the VH:CD in ileum compared to the non-supplementation group. Also, supplementation of 0.05 % STB is significantly lower (P < 0.05) MUC2 and TLR4 gene expression in ileum than the non-supplementation group. At the genus level, the supplementation of 0.05 % STB with NE challenge significantly decreased (P < 0.05) the abundance of Muribaculaceae compared to the NE-challenged group on d 21. In conclusion, supplementation of 0.05 % STB in a diet could positively regulate the cecal microflora and gene expression of tight junction protein and alleviate the decline in nutrient digestibility caused by NE.
Collapse
Affiliation(s)
- Seyeon Chang
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea
| | - Minho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, South Korea
| | - Jihwan Lee
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea Cherrybro Co., Ltd. Jincheon-gun, South Korea 27820
| | - Seonwoong Kim
- Department of Agricultural Economics, Chungbuk National University, Cheongju 28644, South Korea
| | - Dongcheol Song
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea
| | - Kyeongho Jeon
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea
| | - Hyuck Kim
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea
| | - Jinmo Yang
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea
| | - Hyeunbum Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, South Korea.
| | - Jinho Cho
- Department of Animal Science, Chungbuk National University, Cheongju 28644, South Korea.
| |
Collapse
|
7
|
Malczak I, Gajda A, Jedziniak P. Deoxynivalenol and pigs: review of harmful effect of Mycotoxin on swine health. Porcine Health Manag 2025; 11:27. [PMID: 40349078 PMCID: PMC12066055 DOI: 10.1186/s40813-025-00441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Mycotoxins are compounds produced by certain types of fungi, and the mycotoxin one of the most most frequently found in the tested cereal samples is deoxynivalenol (DON), naturally-occurring mycotoxin produced by Fusarium spp. An animal sensitive to the effects of this mycotoxin is the pig due to the diet containing primarily cereals and the structure of a digestive system, which causes DON to be quickly absorbed unchanged into the bloodstream and partially metabolized in the liver. An important aspect when considering the toxicity of DON is the occurrence of its modified forms, which can be transformed into DON in the digestive system. The toxic effect of DON can also be caused by other mycotoxins which co-occur in cereals. The toxic effect of DON on the body of pigs was observed mainly in the digestive, immune, and reproductive systems. The noticeable of DON's toxic effects depends on the exposure time, route of administration and mycotoxin concentration. The changes are mainly caused by impaired gene expression, inhibiting protein synthesis or the effect of DON on pathways in pigs' bodies. The negative impact of DON on the health of pigs may lead to reduced weight gain, poor health, and increased susceptibility to infections and reproductive disorders. There have also been many methods of DON elimination from cereals, but their efficiency is insufficient.
Collapse
Affiliation(s)
- Izabela Malczak
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland.
| | - Anna Gajda
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland
| | - Piotr Jedziniak
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland
| |
Collapse
|
8
|
Feng X, Zhang M, Zhao T, Cui J, Ye H, Zhou C, Ye L, Zhou L. Polystyrene microplastics trigger colonic inflammation in rats via the TLR4/NF-κB/COX-2 pathway and modulation of intestinal microbiota. Toxicology 2025; 513:154090. [PMID: 39971085 DOI: 10.1016/j.tox.2025.154090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Polystyrene microplastics (PS-MPs) are common microplastics that pose significant health hazards to humans. Due to multifunctionality in the gut system, MP-associated damage and mechanisms require further exploration. This study was undertaken with the objective of elucidating the impact of PS-MP exposure on colonic inflammation in rats, and to explore its potential mechanisms. Forty-eight specific-pathogen-free Wistar male rats were administered 0, 0.5, 5, and 50 mg/kg/d of PS-MPs for 90 days, after which intestinal flora distribution, inflammatory factor levels in the colon, and TLR4/NF-κB/COX-2 gene levels were examined. To clarify whether PS-MPs directly infiltrate intestinal epithelial cells and induce cytotoxicity, human intestinal epithelial cells (HIECs) were exposed to a range of PS-MP concentrations (0 ∼ 100 μg/mL) for 48 h, and CCK-8 assays were conducted to assess the cell survival rates. In the colon tissue of rats exposed to PS-MP, goblet cells decreased, muscular layer arrangements were disordered, and disrupted and discontinuous crypt structures appeared in colon tissue, while high numbers of inflammatory cells infiltrated the colonic mucosa and submucosa. PS-MPs could accumulate in HIECs, and cell survival rates were decreased. In the colons of rats exposed to PS-MPs, the levels of Interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α were found to be elevated. Additionally, the mRNA and protein levels of TLR4/MyD88 in the colons of PS-MP-exposed rats exhibited a significant increase. Furthermore, the TLR4/NF-κB/COX-2 signaling pathway in rat colons was activated after MP exposure. When the TLR4/NF-κB/COX-2 signaling pathway was inhibited, the significant increases in IL-6 and TNF-α levels caused by PS-MPs were significantly reversed. PS-MP exposure also altered intestinal flora abundance in rats. Compared with the control group, the proportion of Firmicutes, Proteobacteria and Actinobacteria in PS-MPs exposed group was increased. In contrast, the proportion of Bacteroidetes and Verrucomicrobia decreased. Taken together, our results suggest that PS-MP could exert adverse effects on the gastrointestinal health of rats. Pro-inflammatory cytokine (IL-6, IL-1β and TNF-α) levels increased, and the TLR4/NF-κB/COX-2 signaling pathway was triggered. Thus, flora changes and increased intestinal inflammation may interact with each other.
Collapse
Affiliation(s)
- Xuemin Feng
- Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Meng Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Jianwei Cui
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Hui Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Chunkui Zhou
- Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Azzam MM, Chen W, Xia W, Alagawany M, Elnesr SS, Alabdullatif AA, Alhotan RA, Aboragah AA, Zheng C. Effects of maternal dietary threonine concentrations on the productive performance, amino acid profile in plasma and eggs of laying duck breeders, and performance of ducklings one-week post-hatching. Poult Sci 2025; 104:105031. [PMID: 40121756 PMCID: PMC11981722 DOI: 10.1016/j.psj.2025.105031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
This study's aim was to assess how l- threonine (Thr) affected duck breeders' reproductive and productive performance by examining how it affected the amino acid composition of their eggs and plasma and how well ducklings performed a week after hatching. A total of 648 Longyan duck breeders (23 wk) were randomly allotted to six groups with six replicates of 18 ducks. Ducks were fed a basal control diet deficient in Thr and dietary treatments consisted of the basal diet supplemented l-Thr at 0.0 %, 0.07 %, 0.14 %, 0.21 %, 0.28 %, and 0.35 %, constituting total Thr content of 0.41 %, 0.45 %, 0.51 %, 0.60 %, 0.66 %, and 0.72 %, respectively. At 43 wk of age, the addition of l-Thr at a concentration of 0.28 % had superior (P < 0.05) values (P < 0.05) for egg production, egg weight, egg weight, egg mass, feed conversion ratio, yolk weight and its ratio to total egg weigh compared to the control diet, which yielded the lowest values. Over the entire experimental period, feed intake was not affected by dietary treatments. Plasma tyrosine elevated (P = 0.03 and quadratic trend = 0.02) at 0.28 % l-Thr. In addition, plasma citrulline increased linearly (P = 0.06, linear trend P = 0.003) at 0.28 % l-Thr as compared to the control diet. The contents of aspartic acid, Thr, tyrosine, and proline in egg yolk exhibited a linear increase (P < 0.05) corresponding to increased l- Thr levels. In addition, the contents of serine and cysteine were significantly enhanced at 0.35 % l-Thr, whereas the content of glycine experienced a decrease (P < 0.05) as l- Thr levels increased. The amino acid profile in the egg albumen demonstrated a reduction at 0.28 % and 0.35 % l-Thr, whereas feeding l-Thr at 0.21 % led to an increase in proline contents (P = 0.03, quadratic trend P = 0.002). The addition of l-Thr at a concentration of 0.28 % recorded the heaviest body weight of ducklings after hatching, while the control or 0.07 % Thr group recorded the lowest values. These findings suggest that l-Thr supplementation at 0.28 % (constituting total Thr content of 0.66 %) is an effective nutritional strategy to optimize the performance of duck breeders and the quality of their offspring, providing valuable insights for dietary formulations in poultry production and emphasizing the importance of balanced amino acid nutrition for maximizing breeder performance and offspring quality.
Collapse
Affiliation(s)
- Mahmoud M Azzam
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Weiguang Xia
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| | - Shaaban S Elnesr
- Department of Poultry Production, Faculty of Agriculture, Fayoum University, 63514 Fayoum, Egypt
| | - Abdulaziz A Alabdullatif
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashed A Alhotan
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmad A Aboragah
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Chuntian Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| |
Collapse
|
10
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [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: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
Burton AM, Else KJ, Irving J, Mair I, Shultz S. Antibodies and Inflammation: Fecal Biomarkers of Gut Health in Domestic Ruminants. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2025; 343:468-479. [PMID: 39840509 PMCID: PMC11959687 DOI: 10.1002/jez.2896] [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: 09/25/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025]
Abstract
Gastrointestinal infections present major challenges to ruminant livestock systems, and gut health is a key constraint on fitness, welfare, and productivity. Fecal biomarkers present opportunities to monitor animal health without using invasive methods, and with greater resolution compared to observational metrics. Here we developed enzyme-linked immunosorbent assays for three potential fecal biomarkers of gut health in domestic ruminants: two immunological (total immunoglobulin [Ig]A and total IgG) and one inflammatory (lactoferrin). We analytically validated the assays, then evaluated whether they could be used as a biomarker of clinically diagnosed gastrointestinal pathologies in cattle (Bos taurus), and finally compared them with helminth fecal egg counts in sheep (Ovis aries). The analytes were detected above the lower limits of detection in cattle, sheep, and goats. Fecal IgA and lactoferrin were higher in cattle with infectious pathologies (strongyles, coccidiosis and symptomatic Johne's disease) compared to healthy controls. Lactoferrin was additionally higher in animals with infectious pathologies compared to noninfectious pathologies, and to asymptomatic Johne's cases. No significant relationships were found with sheep fecal egg counts. These initial findings suggest that fecal IgA and lactoferrin may be useful biomarkers of poor gastrointestinal health in cattle, and that fecal lactoferrin is specific to active inflammation caused by infectious agents. These could be incorporated into the growing suite of noninvasive ecoimmunological tools and used to understand ruminant gut health in a range of species. Applications include improving treatment regimens for gastrointestinal infections, and understanding wildlife physiological responses to infectious challenges.
Collapse
Affiliation(s)
- A. M. Burton
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and EngineeringThe University of ManchesterManchesterUK
| | - K. J. Else
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - J. Irving
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and EngineeringThe University of ManchesterManchesterUK
| | - I. Mair
- Institute of Ecology and Evolution, Institute of Immunology and Infection Research, School of Biological SciencesThe University of EdinburghEdinburghUK
| | - S. Shultz
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and EngineeringThe University of ManchesterManchesterUK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| |
Collapse
|
12
|
Szkopek D, Mendel M, Kinsner M, Ognik K, Szyryńska N, Lewczuk B, Kozłowski K, Kos I, Konieczka P. Cannabidiol and nano-selenium mediate intestinal barrier function by affecting mucosal microstructures, and gut-associated immunological and oxidative stress response in the gut of chickens infected with C. perfringens. Front Immunol 2025; 16:1529449. [PMID: 40356900 PMCID: PMC12066498 DOI: 10.3389/fimmu.2025.1529449] [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: 11/16/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Nutritional additives with biological activity, such as cannabidiol (CBD) and nano-selenium (nano-Se), are viable to prevent bacterial diseases such as necrotic enteritis in chickens. The present study hypothesized that CBD and nano-Se mediate epigenetic and oxidative DNA changes in blood and intestinal epithelial cells and can affect intestinal development and functionality in broiler chickens at an early stage of infection with C. perfringens. This study revealed that both compounds, in combination under physiological or pathophysiological conditions, can act synergistically, improving the indices of histomorphometry of duodenum, jejunum, and ileum. Examination of the structures and ultrastructures of the gastrointestinal tract showed that CBD + nano-Se supplementation did not manifest adverse effects on the host gut indices. In contrast, epigenetic and oxidative markers of blood and gut structures indicated that these components balanced the immune system, mitigating the excessive inflammatory response caused by infection, which boosted the immune response of birds to challenge. There were also significant correlations between indicators of intestinal barrier function, such as diamine oxidase and lactic acid levels, and histomorphometry and markers of DNA integrity in the blood and intestine of chickens. In addition, it was shown that nano-Se increased hemoglobin concentration, which may be beneficial in the host's response to pathogen stimuli. These findings evidenced the health-promoting effect of cannabidiol and nano-selenium in C. perfringens-infected chickens and provided new insights into the mechanism of action of both nutritional additives.
Collapse
Affiliation(s)
- Dominika Szkopek
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Marta Mendel
- Division of Pharmacology and Toxicology, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Misza Kinsner
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Katarzyna Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bio-Economy, University of Life Sciences in Lublin, Lublin, Poland
| | - Natalia Szyryńska
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Bogdan Lewczuk
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Krzysztof Kozłowski
- Department of Poultry Science and Apiculture, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Ivica Kos
- Department of Animal Science and Technology, Faculty of Agriculture, University of Zagreb, Zagreb, Croatia
| | - Paweł Konieczka
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
- Department of Poultry Science and Apiculture, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
13
|
Ladaika CA, Chakraborty A, Masood A, Hostetter G, Yi JM, O'Hagan HM. LSD1 inhibition attenuates targeted therapy-induced lineage plasticity in BRAF mutant colorectal cancer. Mol Cancer 2025; 24:122. [PMID: 40264166 PMCID: PMC12016338 DOI: 10.1186/s12943-025-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND BRAF activating mutations occur in approximately 10% of metastatic colorectal cancer (CRCs) and are associated with worse prognosis in part due to an inferior response to standard chemotherapy. Standard of care for patients with refractory metastatic BRAFV600E CRC is treatment with BRAF and EGFR inhibitors and recent FDA approval was given to use these inhibitors in combination with chemotherapy for patients with treatment naïve metastatic BRAFV600E CRC. Lineage plasticity to neuroendocrine cancer is an emerging mechanism of targeted therapy resistance in several cancer types. Enteroendocrine cells (EECs), the neuroendocrine cell of the intestine, are uniquely present in BRAF mutant CRC as compared to BRAF wildtype CRC. METHODS BRAF plus EGFR inhibitor treatment induced changes in cell composition were determined by gene expression, imaging and single cell approaches in multiple models of BRAF mutant CRC. Furthermore, multiple clinically relevant inhibitors of the lysine demethylase LSD1 were tested to determine which inhibitor blocked the changes in cell composition. RESULTS Combined BRAF and EGFR inhibition enriched for EECs in all BRAF mutant CRC models tested. Additionally, EECs and other secretory cell types were enriched in a subset of BRAFV600E CRC patient samples following targeted therapy. Importantly, inhibition of LSD1 with a clinically relevant inhibitor attenuated targeted therapy-induced EEC enrichment through blocking the interaction of LSD1, CoREST2 and STAT3. CONCLUSIONS Our findings that BRAF plus EGFR inhibition induces lineage plasticity in BRAFV600E CRC represents a new paradigm for how resistance to BRAF plus EGFR inhibition occurs. Additionally, our finding that LSD1 inhibition blocks lineage plasticity has the potential to improve responses to BRAF plus EGFR inhibitor therapy in patients.
Collapse
Affiliation(s)
- Christopher A Ladaika
- Genome, Cell, and Developmental Biology Graduate Program, Department of Biology, Indiana University Bloomington, Bloomington, IN, 47405, USA
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
| | - Averi Chakraborty
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
| | - Ashiq Masood
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Joo Mi Yi
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, South Korea
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Heather M O'Hagan
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA.
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine, Bloomington, IN, 47405, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
14
|
Subramanian D, Chin A, Shi Y, Liu GW, Langer R, Traverso G. Identification and Validation of Cyclic Peptides with Mucin-Selective, Location-Specific Binding in the Gastrointestinal Tract. ACS NANO 2025; 19:14693-14706. [PMID: 40216380 PMCID: PMC12020424 DOI: 10.1021/acsnano.4c13520] [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: 09/25/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025]
Abstract
Oral drug delivery is a widely preferred method of drug administration due to its ease of use and convenience for patients. Localization of drug release in the gastrointestinal (GI) tract is important to treat localized diseases and maximize drug absorption. However, achieving drug localization in the dynamic GI tract is challenging. To address this challenge, we leveraged the geographic diversity of the GI tract by targeting its mucus layers, which coat the epithelial surfaces. These layers, composed of mucin glycoproteins, are synthesized with unique chemical compositions and expressed in different regions, making them ideal targets for drug localization. In this article, we identify cyclic peptides that bind selectively to MUC2 (in the intestines) and MUC5AC (in the stomach), serving as targeting ligands to these regions of the GI tract. We demonstrate the effectiveness of these peptides through in vitro, ex vivo, and in vivo experiments, showing that incorporating these targeting ligands can increase binding and selectivity 2-fold to the desired regions, thus potentially overcoming challenges with localizing drug distribution in oral delivery. These results indicate that cyclic peptides can be used to localize drug cargoes at certain sites in the body compared to free drugs.
Collapse
Affiliation(s)
- Deepak
A. Subramanian
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Austin Chin
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Yunhua Shi
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Gary W. Liu
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Langer
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Giovanni Traverso
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
15
|
Dash SK, Marques CNH, Mahler GJ. Small Intestine on a Chip Demonstrates Physiologic Mucus Secretion in the Presence of Lacticaseibacillus rhamnosus Biofilm. Biotechnol Bioeng 2025. [PMID: 40197633 DOI: 10.1002/bit.28989] [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: 11/20/2024] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
The small intestine is an area of the digestive system difficult to access using current medical procedures, which prevents studies on the interactions between food, drugs, the small intestinal epithelium, and resident microbiota. Therefore, there is a need to develop novel microfluidic models that mimic the intestinal biological and mechanical environments. These models can be used for drug discovery and disease modeling and have the potential to reduce reliance on animal models. The goal of this study was to develop a small intestine on a chip with both enterocyte (Caco-2) and goblet (HT29-MTX) cells cocultured with Lacticaseibacillus rhamnosus biofilms, which is of one of several genera present in the small intestinal microbiota. L. rhamnosus was introduced following the establishment of the epithelial barrier. The shear stress within the device was kept in the lower physiological range (0.3 mPa) to enable biofilm development over the in vitro epithelium. The epithelial barrier differentiated after 5 days of dynamic culture with cell polarity and permeability similar to the human small intestine. The presence of biofilms did not alter the barrier's permeability in dynamic conditions. Under fluid flow, the complete model remained viable and functional for more than 5 days, while the static model remained functional for only 1 day. The presence of biofilm increased the secretion of acidic and neutral mucins by the epithelial barrier. Furthermore, the small intestine on a chip also showed increased MUC2 production, which is a dominant gel-forming mucin in the small intestine. This model builds on previous publications as it establishes a stable environment that closely mimics in vivo conditions and can be used to study intestinal physiology, food-intestinal interactions, and drug development.
Collapse
Affiliation(s)
- Sanat Kumar Dash
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Cláudia N H Marques
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
16
|
Subramanian DA, Kirtane AR, White GN, Freitas DE, Ishida K, Jenkins J, Pettinari A, Morimoto J, Fitzgerald N, Traverso G. Identification and Validation of Small Molecules with Mucin-Selective Regiospecific Binding in the Gastrointestinal Tract. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646052. [PMID: 40235971 PMCID: PMC11996415 DOI: 10.1101/2025.03.31.646052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Oral drug delivery is a widely used method of drug administration; however, achieving localized drug release at specific regions of the gastrointestinal (GI) tract is generally accomplished by using broad environmental differences. The GI tract is a complex system with regional differences in composition, such as selective expression of mucin glycoproteins in different organs. Here, we identify small molecule ligands that can selectively bind to the different mucins to localize drug delivery to the small intestine and stomach. We demonstrate up to a 10-fold increase in particle binding to these organs and up to a 4-fold increase in selectivity compared to chitosan. Additionally, we observe up to a 9-fold increase in budesonide concentration in the small intestine and a 25-fold increase in tetracycline concentration in the stomach. These results show that we have developed a versatile platform capable of sequestering a variety of drugs in certain GI tract organs.
Collapse
|
17
|
Yousefi Y, Haider Z, Grondin JA, Wang H, Haq S, Banskota S, Seto T, Surette M, Khan WI. Gut microbiota regulates intestinal goblet cell response and mucin production by influencing the TLR2-SPDEF axis in an enteric parasitic infection. Mucosal Immunol 2025:S1933-0219(25)00033-9. [PMID: 40164286 DOI: 10.1016/j.mucimm.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Alterations in goblet cell biology constitute one of the most effective host responses against enteric parasites. In the gastrointestinal (GI) tract, millions of bacteria influence these goblet cell responses by binding to pattern recognition receptors such as toll-like receptors (TLRs). Studies suggest that the gut microbiota also interacts bidirectionally with enteric parasites, including Trichuris muris. Here, we study the roles of T. muris-altered microbiota and the TLR2-SPDEF axis in parasitic host defense. In acute T. muris infection, we observed altered gut microbiota composition, which, when transferred to germ-free mice, resulted in increased goblet cell numbers, Th2 cytokines and Muc2 expression, as well as increased Tlr2. Further, antibiotic (ABX)-treated TLR2-/- mice, despite having received the same T. muris-altered microbiota, displayed diminished Th2 response, Muc2 expression, and, intriguingly, diminished SPDEF expression compared to wildtype counterparts. When infected with T. muris, SPDEF-/- mice exhibited a reduced Th2 response and altered microbial composition compared to SPDEF+/+, particularly on day 14 post-infection, and this microbiota was sufficient to alter host goblet cell response when transferred to ABX-treated mice. Taken together, our findings suggest the TLR2-SPDEF axis, via T. muris-induced microbial changes, is an important regulator of goblet cell function and host's parasitic defense.
Collapse
Affiliation(s)
- Yeganeh Yousefi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Zarin Haider
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jensine A Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Huaqing Wang
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sabah Haq
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Suhrid Banskota
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Tyler Seto
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Michael Surette
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
18
|
Almawash S. Oral Bioavailability Enhancement of Anti-Cancer Drugs Through Lipid Polymer Hybrid Nanoparticles. Pharmaceutics 2025; 17:381. [PMID: 40143044 PMCID: PMC11946161 DOI: 10.3390/pharmaceutics17030381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/13/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer is considered as the second leading cause of death worldwide. Chemotherapy, radiotherapy, immunotherapy, and targeted drug delivery are the main treatment options for treating cancers. Chemotherapy drugs are either available for oral or parenteral use. Oral chemotherapy, also known as chemotherapy at home, is more likely to improve patient compliance and convenience. Oral anti-cancer drugs have bioavailability issues associated with lower aqueous solubility, first-pass metabolism, poor intestinal permeability and drug absorption, and degradation of the drug throughout its journey in the gastrointestinal tract. A highly developed carrier system known as lipid polymer hybrid nanoparticles (LPHNs) has been introduced. These nanocarriers enhance drug stability, solubility, and absorption, and reduce first-pass metabolism. Consequently, this will have a positive impact on oral bioavailability enhancement. This article provides an in-depth analysis of LPHNs as a novel drug delivery system for anti-cancer agents. It discusses an overview of the limited bioavailability of anti-cancer drugs, their reasons and consequences, LPHNs based anti-cancer drug delivery, conventional and modern preparation methods as well as their drug loading and entrapment efficiencies. In addition, this article also gives an insight into the mechanistic approach to oral bioavailability enhancement, potential applications in anti-cancer drug delivery, limitations, and future prospects of LPHNs in anti-cancer drug delivery.
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
19
|
Moraitis I, Taelman J, Arozamena B, Mularoni L, Wienskowska O, Sanjuan Garriga X, Arregui L, Stefanovic M, Modolell Farré I, Guedea F, Diaz M, Guiu J. Mucosal Macrophages Govern Intestinal Regeneration in Response to Injury. Gastroenterology 2025:S0016-5085(25)00465-2. [PMID: 40086603 DOI: 10.1053/j.gastro.2025.01.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/15/2025] [Accepted: 01/30/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND & AIMS Radiation-induced enteritis develops in cancer patients treated with radiotherapy in the abdominal and pelvic cavity, a condition that impairs their quality of life. Radiation injury depletes proliferative intestinal stem cells; in response to this, the epithelium activates a regenerative program that facilitates the healing of the intestine. However, the mechanisms that induce the activation of the intestinal regenerative program are poorly characterized. METHODS In this study, we induced radiation-induced enteritis in mice through abdominal irradiation, mimicking clinical scenarios. Through imaging and flow cytometric analysis, we investigated the recruitment of macrophages to the small intestine during injury and healing. Additionally, we developed a coculture system for mouse and human intestinal organoids and macrophages to explore the cross talk between these cells. Then by combining in vivo ablation of macrophages, fluorescent lineage tracing, imaging, bulk RNA-sequencing (RNA-seq), single-cell RNA-seq, human intestinal organoids, and cell trajectory analysis, we studied the macrophage induction of intestinal regeneration at the cellular and molecular level. RESULTS Our findings revealed that macrophages are recruited around the intestinal stem cell compartment upon radiation injury, promoting a fetal-like reprogramming and proliferation of epithelial cells that drives the regeneration process. In contrast, macrophage ablation led to compromised regeneration. Moreover, our single-cell RNA-seq analysis identified key secreted molecules, neuregulin 1 and osteopontin, as pivotal players in regulating this process. Additionally, characterization of human macrophage/organoid cocultures and cell trajectory inference confirmed the conservation of macrophages' role in triggering the regenerative program in primary human cells. CONCLUSIONS This study identifies macrophages as essential contributors to intestinal regeneration beyond their innate immune response. Targeting macrophages therapeutically may hold promise in enhancing regeneration and improving the quality of life for cancer survivors.
Collapse
Affiliation(s)
- Ilias Moraitis
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain; Biomedicine PhD Program, Universitat de Barcelona, Barcelona, Spain
| | - Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Borja Arozamena
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Loris Mularoni
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Olga Wienskowska
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Xavier Sanjuan Garriga
- Department of Pathology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Laura Arregui
- HUB-ICO-IDIBELL Biobank, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Milica Stefanovic
- Department of Radiobiology and Cancer, ONCOBELL, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Radiation Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ignasi Modolell Farré
- Servei de Física Mèdica i Protecció Radiològica, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ferran Guedea
- Department of Radiobiology and Cancer, ONCOBELL, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Department of Radiation Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mònica Diaz
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona, Spain; Centre for Networked Biomedical Research on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
20
|
Pohl J, Aretakis D, Tacke F, Engelmann C, Sigal M. Role of Intestinal Barrier Disruption to Acute-on-Chronic Liver Failure. Semin Liver Dis 2025; 45:52-65. [PMID: 40081417 DOI: 10.1055/a-2516-2361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Acute-on-chronic liver failure (ACLF) is a severe condition in patients with decompensated liver cirrhosis, marked by high short-term mortality. Recent experimental and clinical evidence has linked intestinal dysfunction to both the initiation of ACLF as well as disease outcome. This review discusses the significant role of the gut-liver axis in ACLF pathogenesis, highlighting recent advances. Gut mucosal barrier disruption, gut dysbiosis, and bacterial translocation emerge as key factors contributing to systemic inflammation in ACLF. Different approaches of therapeutically targeting the gut-liver axis via farnesoid X receptor agonists, nonselective beta receptor blockers, antibiotics, and probiotics are discussed as potential strategies mitigating ACLF progression. The importance of understanding the distinct pathophysiology of ACLF compared with other stages of liver cirrhosis is highlighted. In conclusion, research findings suggest that disruption of intestinal integrity may be an integral component of ACLF pathogenesis, paving the way for novel diagnostic and therapeutic approaches to manage this syndrome more effectively.
Collapse
Affiliation(s)
- Julian Pohl
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dimitrios Aretakis
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Cornelius Engelmann
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
21
|
Ayala C, Sathe A, Bai X, Grimes SM, Shen J, Poultsides GA, Lee B, Ji HP. Distinct gene signatures define the epithelial cell features of mucinous appendiceal neoplasms and pseudomyxoma metastases. Front Genet 2025; 16:1536982. [PMID: 40018643 PMCID: PMC11865047 DOI: 10.3389/fgene.2025.1536982] [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: 11/29/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Appendiceal mucinous neoplasms (AMN) are rare tumors of the gastrointestinal tract. They metastasize with widespread abdominal dissemination leading to pseudomyxoma peritonei (PMP), a disease with poor prognosis. There are many unknowns about the cellular features of origin, differentiation and progression of AMN and PMP. Methods We characterized AMNs, PMPs and matched normal tissues using single-cell RNA-sequencing. We validated our findings with immunohistochemistry, mass spectrometry on malignant ascites from PMP patients and gene expression data from an independent set of PMP tumors. Results We identified previously undescribed cellular features and heterogeneity in AMN and PMP tumors. There were gene expression signatures specific to the tumor epithelial cells among AMN and PMP. These signatures included genes indicative of goblet cell differentiation and elevated mucin gene expression. Metastatic PMP cells had a distinct gene expression signature with increased lipid metabolism, inflammatory, JAK-STAT and RAS signaling pathway among others. We observed clonal heterogeneity in a single PMP tumor as well as PMP metastases from the same patient. Discussion Our study defined tumor cell gene signatures of AMN and PMP, successfully overcoming challenges of low cellularity and mucinous composition of these tumors. These gene expression signatures provide insights on tumor origin and differentiation, together with the identification of novel treatment targets. The heterogeneity observed within an individual tumor and between different tumors from the same patient, represents a potential source of treatment resistance.
Collapse
Affiliation(s)
- Carlos Ayala
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Susan M. Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Jeanne Shen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - George A. Poultsides
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Byrne Lee
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Hanlee P. Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
22
|
Abu-Siniyeh A, Khataibeh M, Al-Zyoud W, Al Holi M. Zebrafish as a model for human epithelial pathology. Lab Anim Res 2025; 41:6. [PMID: 39901304 PMCID: PMC11789318 DOI: 10.1186/s42826-025-00238-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Zebrafish (Danio rerio) have emerged as an influential model for studying human epithelial pathology, particularly because of their genetic similarity to humans and their unique physiological traits. This review explores the structural and functional homology between zebrafish and human epithelial tissues in organs, such as the gastrointestinal system, liver, and kidneys. Zebrafish possess significant cellular and functional homology with mammals, which facilitates the investigation of various diseases, including inflammatory bowel disease, nonalcoholic fatty liver disease, and polycystic kidney disease. The advantages of using zebrafish as a model organism include rapid external development, ease of genetic manipulation, and advanced imaging capabilities, allowing for the real-time observation of disease processes. However, limitations exist, particularly concerning the lack of organs in zebrafish and the potential for incomplete phenocopy of human conditions. Despite these challenges, ongoing research in adult zebrafish promises to enhance our understanding of the disease mechanisms and regenerative processes. By revealing the similarities and differences in epithelial cell function and disease pathways, this review highlights the value of zebrafish as a translational model for advancing our knowledge of human health and developing targeted therapies.
Collapse
Affiliation(s)
- Ahmed Abu-Siniyeh
- Department of Medical Laboratory Sciences, School of Science, The University of Jordan, Amman, Jordan.
| | - Moayad Khataibeh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, As Salt, Jordan
| | - Walid Al-Zyoud
- Department of Biomedical Engineering, School of Applied Medical Sciences, German Jordanian University, Amman, 11180, Jordan
| | - Majed Al Holi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
23
|
Soltani N, Rahimi S, Khaki P, Karimi Torshizi MA, Eskandari B, Grimes J. Efficacy of hyperimmunized egg yolk antibodies (IgY) against Campylobacter jejuni: In Vitro and In Vivo evaluations. Poult Sci 2025; 104:104718. [PMID: 39787828 PMCID: PMC11761915 DOI: 10.1016/j.psj.2024.104718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Campylobacter infections are a prevalent cause of diarrheal disease in humans and are the most significant zoonotic pathogens worldwide. Human campylobacteriosis is generally via ingestion of contaminated poultry products. However, based on recent studies chicken egg yolk antibody (IgY) powder has great potential to reduce the cecum load of Campylobacter jejuni (C. jejuni) in broilers. To understand the effective and economically feasible dosage, two immunization and challenge studies were conducted using 30 layer hens and 250 broiler chickens and found a scientific approach, starting with in vitro evaluations and progressing with in vivo studies confirmed. In this study it was demonstrated that specific IgY powder (SIgY), produced by immunized hens via bacterin, was highly effective in inhibiting bacterial growth and adhesion, as well as exhibiting bactericidal and agglutination properties (P < 0.05). Notably, doses of 0.5 % and 1 % SIgY significantly enhanced both the height and width of intestinal villi, along with improving the villus height-to-crypt depth ratio when compared to the positive control group (P < 0.05). Furthermore, medium and high doses of SIgY were effective in preserving the integrity of the intestinal epithelium, as evidenced by a reduction in crypt depth and the number of goblet cells, which serve as important markers in the immune system (P < 0.01). Additionally, analyses of cecal and liver bacterial counts in response to the 0.5 % SIgY treatment revealed a significant reduction in C. jejuni counts compared to other challenged groups throughout the 28 d experiment (P < 0.01). Based on these results, it may be concluded that specific antibodies play a crucial role in maintaining the integrity of intestinal villi, support the health of the intestinal epithelium, and reduce the colonization of C. jejuni. These findings could form the basis for developing an economical and effective strategy to enhance poultry and human health in the context of C. jejuni infection.
Collapse
Affiliation(s)
- Nazanin Soltani
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Shaban Rahimi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Pejvak Khaki
- Department of Microbiology, Razi Vaccine and Serum Production Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Postal Code: 3197619751, Karaj, Iran
| | - Mohammad Amir Karimi Torshizi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Bahareh Eskandari
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Jesse Grimes
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC 27695-7608.
| |
Collapse
|
24
|
Damianos J, Abdelnaem N, Camilleri M. Gut Goo: Physiology, Diet, and Therapy of Intestinal Mucus and Biofilms in Gastrointestinal Health and Disease. Clin Gastroenterol Hepatol 2025; 23:205-215. [PMID: 39426645 PMCID: PMC11761393 DOI: 10.1016/j.cgh.2024.09.007] [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: 07/24/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The gastrointestinal tract has remarkable capacity to withstand considerable insults from exposure to abrasive food particles, chemicals, allergens, and pathogenic microbes. Maintaining a robust epithelial barrier sequesters these potentially harmful substances in the lumen, preventing absorption into the systemic circulation. Normal functioning of this barrier is central in diverse physiological processes including digestion, immunity, inflammation, and gut-brain signaling. One crucial component of the barrier is the mucus layer covering the epithelium. There is increased appreciation of the importance of mucus in maintenance of the gut barrier, and how dysregulation of the mucus layer contributes to several common gastrointestinal pathologies. This manuscript reviews the physical and chemical properties of mucus, its maintenance and turnover, and its role in maintaining gut barrier integrity. The dynamic interactions of the mucus layer within the gut ecosystem are illustrated by highlighting how a weakened mucus layer or defective mucus production facilitate pathogenic microbial colonization and mucosal biofilm formation. These may potentially contribute to the pathogenesis of gastrointestinal diseases such as inflammatory bowel diseases or result in secretion and mucosal damage and inflammation in bile acid diarrhea. A final goal is to review how certain dietary factors, especially low-fiber diets and emulsifiers common in Western diets, can harm the mucus layer. This report summarizes evidence from preclinical and human studies that document damage to the mucus layer, and reviews approaches, including diets and probiotics, that promote a healthy mucus layer and break down pathogenic biofilms, thereby potentially preventing and/or treating gastrointestinal diseases that impact mucosal integrity.
Collapse
Affiliation(s)
- John Damianos
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Nada Abdelnaem
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
25
|
Jankowski WM, Fichna J, Tarasiuk-Zawadzka A. A systematic review of the relationship between gut microbiota and prevalence of pancreatic diseases. Microb Pathog 2025; 199:107214. [PMID: 39653281 DOI: 10.1016/j.micpath.2024.107214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/24/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Acute pancreatitis (AP) represents one of the most common gastrointestinal (GI) diseases; it can manifest in varying degrees of severity, sometimes leading to a life-threatening condition for the patient. Pancreatic ductal adenocarcinoma (PDAC), due to its high malignancy and uncertain prognosis, is widely regarded as one of the most fatal diseases. The increasing prevalence of AP and PDAC represents a major burden on public health and the healthcare system worldwide. The aim of this systematic review was to discuss the current state of knowledge regarding the relationship between the gut microbiota and the incidence, prognosis, diagnosis and treatment of AP and PDAC. To identify studies that analyzed the relationship between the gut microbiota and the occurrence/development of pancreatic diseases or PDAC, the online databases PubMed, Scopus and Google Scholar were searched between November 2023 and January 2024. Finally, 14 publications met the inclusion criteria (1. were conducted exclusively in humans and/or animals; 2. original, published in English in peer-reviewed journals after 2019; 3. described the relationship between gut microbiota and the occurrence of AP or PDAC). The collected studies indicated significant changes in the gut microbiota of patients with AP and PDAC. Moreover, they highlighted the presence of a relationship between the gut microbiota and the occurrence, course, treatment efficiency and prognosis of the disease in question. Further research is needed to understand precisely the relationship between the gut microbiota and the occurrence of pancreatic diseases and whether it may be a starting point for the development of modern forms of therapy based on the use of prebiotics and/or diet to restore the normal composition of the intestinal bacteria.
Collapse
Affiliation(s)
- Wojciech Michał Jankowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Aleksandra Tarasiuk-Zawadzka
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| |
Collapse
|
26
|
Wan MLY, Co VA, Turner PC, Nagendra SP, El‐Nezami H. Deoxynivalenol modulated mucin expression and proinflammatory cytokine production, affecting susceptibility to enteroinvasive Escherichia coli infection in intestinal epithelial cells. J Food Sci 2025; 90:e70079. [PMID: 39980277 PMCID: PMC11842951 DOI: 10.1111/1750-3841.70079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025]
Abstract
Deoxynivalenol (DON) is a common mycotoxin in crops that could induce intestinal inflammation, affecting the susceptibility of intestinal epithelial cells (IECs) to pathogen infection. This study aimed to investigate DON's effects on mucin and cytokine production as part of the local immune system and how it affected intestinal susceptibility to pathogen infection. Caco-2 cells were exposed to DON followed by acute enteroinvasive Escherichia coli (EIEC) infection. An increase in EIEC attachment to DON-exposed cells was observed, probably in part, mediated by secretory MUC5AC mucins and membrane-bound MUC4 and MUC17 mucins. Additionally, DON with EIEC posttreatment led to significant changes in the gene expression of several proinflammatory cytokines (IL1α, IL1β, IL6, IL8, TNFα, and MCP-1), which may be in part, mediated by NK-κB and/or MAPK signaling pathways. These data suggested DON may exert immunomodulatory effects on IECs, altering the IEC susceptibility to bacterial infection. PRACTICAL APPLICATION: The results suggested that DON might modulate immune responses by affecting mucus and cytokine production, which may affect the susceptibility of intestinal epithelial cells to pathogen infection.
Collapse
Affiliation(s)
- Murphy Lam Yim Wan
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences BuildingThe University of Hong KongPokfulamHong Kong
- Department of Laboratory Medicine, Division of MicrobiologyImmunology and Glycobiology, Lund UniversityLundSweden
- School of Medicine, Pharmacy and Biomedical Sciences, Faculty of Science and HealthUniversity of PortsmouthPortsmouthUK
| | - Vanessa Anna Co
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences BuildingThe University of Hong KongPokfulamHong Kong
| | - Paul C Turner
- Maryland Institute for Applied Environmental Health, School of Public HealthUniversity of MarylandCollege ParkMarylandUSA
| | - Shah P Nagendra
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences BuildingThe University of Hong KongPokfulamHong Kong
| | - Hani El‐Nezami
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences BuildingThe University of Hong KongPokfulamHong Kong
- Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| |
Collapse
|
27
|
Calvez V, Puca P, Di Vincenzo F, Del Gaudio A, Bartocci B, Murgiano M, Iaccarino J, Parand E, Napolitano D, Pugliese D, Gasbarrini A, Scaldaferri F. Novel Insights into the Pathogenesis of Inflammatory Bowel Diseases. Biomedicines 2025; 13:305. [PMID: 40002718 PMCID: PMC11853239 DOI: 10.3390/biomedicines13020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs), encompassing Crohn's disease and ulcerative colitis, are complex chronic disorders characterized by an intricate interplay between genetic predisposition, immune dysregulation, gut microbiota alterations, and environmental exposures. This review aims to synthesize recent advances in IBD pathogenesis, exploring key mechanisms and potential avenues for prevention and personalized therapy. A comprehensive literature search was conducted across major bibliographic databases, selecting the most recent and impactful studies on IBD pathogenesis. The review integrates findings from multi-omics analyses, single-cell transcriptomics, and longitudinal cohort studies, focusing on immune regulation, gut microbiota dynamics, and environmental factors influencing disease onset and progression. Immune dysregulation, including macrophage polarization (M1 vs. M2) and Th17 activation, emerges as a cornerstone of IBD pathogenesis. Dysbiosis, as a result of reduced alpha and beta diversity and overgrowth of harmful taxa, is one of the main contributing factors in causing inflammation in IBD. Environmental factors, including air and water pollutants, maternal smoking, and antibiotic exposure during pregnancy and infancy, significantly modulate IBD risk through epigenetic and microbiota-mediated mechanisms. While recent advances have supported the development of new therapeutic strategies, deeply understanding the complex dynamics of IBD pathogenesis remains challenging. Future efforts should aim to reduce the burden of disease with precise, personalized treatments and lower the incidence of IBD through early-life prevention and targeted interventions addressing modifiable risk factors.
Collapse
Affiliation(s)
- Valentin Calvez
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Pierluigi Puca
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Federica Di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Jacopo Iaccarino
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Erfan Parand
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Daniele Napolitano
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
| | - Daniela Pugliese
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Franco Scaldaferri
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| |
Collapse
|
28
|
Barnabé MLDF, Vicente LCDOS, Martins KVC, Lacerda GF, Rodrigues E, Oliveira LA, Dias KA, Pereira SMS, José VPBDS, Dias MMDS, Calhelha RC, Leite LB, Ribeiro L, de Carvalho IMM, da Silva BP, Martino HSD, Gonçalves RV, Mattos Della Lucia C. Soybean Flour Fortified with Gryllus assimilis Powder to Increase Iron Bioavailability Improves Gut Health and Oxidative Balance In Vivo. Nutrients 2025; 17:437. [PMID: 39940294 PMCID: PMC11819770 DOI: 10.3390/nu17030437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Insects like Gryllus assimilis have an excellent nutritional profile, including iron. However, the bioavailability of this iron and its effects on intestinal health and oxidative balance remain unclear. To enhance acceptance, insects can be used in powder form and combined with common flours. OBJECTIVE This study evaluates the effects of Gryllus assimilis powder, alone or with soy flour, on iron bioavailability, intestinal health, and oxidative balance in rodents. METHODS Using the hemoglobin depletion/repletion method, 32 male Wistar rats were divided into four groups: A (standard diet + ferrous sulfate), B (diet + Gryllus assimilis + soy flour), C (diet + Gryllus assimilis), and D (diet + soy flour). Hemoglobin levels, regeneration efficiency, biological value, serum markers, intestinal health, and oxidative balance were assessed. RESULTS Food intake, weight gain, and bioavailability measures showed no differences. However, the Gryllus + soy group showed higher weekly and final hemoglobin levels than Gryllus alone. This combination also improved acetic acid levels, fecal moisture, and oxidative balance, increasing superoxide dismutase activity while reducing peroxidation products compared to Gryllus alone. CONCLUSION These findings highlight the potential benefits of combining Gryllus assimilis with soy flour for iron bioavailability and overall health.
Collapse
Affiliation(s)
- Michele Lílian da Fonseca Barnabé
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Laura Célia de Oliveira Souza Vicente
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Karina Vitoria Cipriana Martins
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Gabrieli Fernandes Lacerda
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Elias Rodrigues
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Lívya Alves Oliveira
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Kelly Aparecida Dias
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Stephanie Michelin Santana Pereira
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| | - Vinicius Parzanini Brilhante de São José
- Laboratory of Experimental Nutrition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (V.P.B.d.S.J.); (B.P.d.S.); (H.S.D.M.)
| | | | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Alameda Santa Apolónia, 5300-252 Bragança, Portugal;
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-252 Bragança, Portugal
| | - Luciano Bernardes Leite
- Department of Physical Education, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil;
- Department of Sports, Instituto Politécnico de Bragança, 5300-252 Bragança, Portugal
| | - Lúcia Ribeiro
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Alameda Santa Apolónia, 5300-252 Bragança, Portugal;
- Laboratório Associado para a Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-252 Bragança, Portugal
- Facultade de Ciencias, Universidad de Vigo, 32004 Ourense, Spain
| | | | - Bárbara Pereira da Silva
- Laboratory of Experimental Nutrition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (V.P.B.d.S.J.); (B.P.d.S.); (H.S.D.M.)
| | - Hércia Stampini Duarte Martino
- Laboratory of Experimental Nutrition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (V.P.B.d.S.J.); (B.P.d.S.); (H.S.D.M.)
| | - Reggiani Vilela Gonçalves
- Department of Animal Biology, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.M.d.S.D.); (R.V.G.)
| | - Ceres Mattos Della Lucia
- Laboratory of Vitamin Analysis, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (M.L.d.F.B.); (L.C.d.O.S.V.); (K.V.C.M.); (G.F.L.); (E.R.); (L.A.O.); (K.A.D.); (S.M.S.P.); (C.M.D.L.)
| |
Collapse
|
29
|
Huang MY, Smieska LM, Tako E. Intestinal Effects of Brewers' Spent Grain Extract In Ovo ( Gallus gallus)-A Pilot Study. Animals (Basel) 2025; 15:303. [PMID: 39943073 PMCID: PMC11816252 DOI: 10.3390/ani15030303] [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: 12/19/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Upcycling brewers' spent grain (BSG) into poultry feed needs to be optimized. Since broiler chickens inefficiently digest fiber, we created a water-soluble BSG extract (BSGE) to explore this fraction's potential nutritional benefits. We utilized intra-amniotic administration (in ovo) to target the gastrointestinal tract of broiler embryos. BSGE increased villus surface area and goblet cell quantity and size, implying improved duodenal development. The extract also changed cecal Escherichia coli (E. coli) and Clostridium abundances. Synchrotron X-ray fluorescence microscopy, along with zinc and iron transporter relative expression, did not reveal significant changes by BSGE. These findings highlight the potential for BSGE to be a functional feed component, underscoring the potential value of upcycling this byproduct. This pilot study supports future work exploring the impact of BSGE within feed and its effects over long-term consumption.
Collapse
Affiliation(s)
- Melissa Y. Huang
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA;
| | - Louisa M. Smieska
- Cornell High Energy Synchrotron Source, Cornell University, Ithaca, NY 14853, USA;
| | - Elad Tako
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
30
|
D’Alessandro AG, Di Luca A, Desantis S, Martemucci G. Antioxidant Synergy in a Mixture of Powder Plant Leaves and Effects on Metabolic Profile, Oxidative Status and Intestinal Morpho-Histochemical Features of Laying Hens. Animals (Basel) 2025; 15:308. [PMID: 39943078 PMCID: PMC11816074 DOI: 10.3390/ani15030308] [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: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Phenolic antioxidant intake is encouraged to prevent oxidative damage, and antioxidant synergy is considered an advantage in adding polyphenols from varied plants. This study investigated the antioxidant and synergistic interactions among olive leaf (OL), bay laurel (BL), and rosemary (RL) leaf powder mixture (LPM: OL + BL + RL), using in vitro chemical tests [TPC, ORAC, TEAC-ABTS, FRAP; combination index (CI)], and in vivo validation on blood oxidative status, metabolic profile, and intestinal histomorphology in laying hens. The in vitro study indicated a whole higher antioxidant capacity for the LPM than respective single/double-leave combinations. The LPM CI value (IC50, 0.60) indicated a synergistic effect compared to the binary combinations. Thus, the LPM was validated in vivo through dietary supplementation on sixty Lohmann Brown hens (30 weeks old), reared in an indoor-outdoor rearing system divided. The hens were allocated into two experimental groups (n. 30): basal control diet group; and diet supplemented group with 6 g/kg feed of LPM) containing OL, BL, and RL (respectively, at 65.7%:18.9%:15.4%), for 60 days. The LPM improved (p < 0.05) the oxidative status (TAS, FRAP; ROMs, TBARs) and vitamin E level, metabolic and immunological profiles, and it induced region-specific changes in the morphology and carbohydrate composition of mucins along intestinal tracts of the animals. These findings could provide a valuable strategy for identifying synergistic combinations in functional feed formulations for laying hens.
Collapse
Affiliation(s)
- Angela Gabriella D’Alessandro
- Department of Soil, Plant and Food Sciences (DiSSPA), University of Bari Aldo Moro, 70126 Bari, Italy; (A.G.D.); (A.D.L.); (G.M.)
| | - Alessio Di Luca
- Department of Soil, Plant and Food Sciences (DiSSPA), University of Bari Aldo Moro, 70126 Bari, Italy; (A.G.D.); (A.D.L.); (G.M.)
| | - Salvatore Desantis
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giovanni Martemucci
- Department of Soil, Plant and Food Sciences (DiSSPA), University of Bari Aldo Moro, 70126 Bari, Italy; (A.G.D.); (A.D.L.); (G.M.)
| |
Collapse
|
31
|
Chen K, Wang L, Liu J, Zheng H, Wu X, Liao X. The ant that may well destroy a whole dam: a systematic review of the health implication of nanoplastics/microplastics through gut microbiota. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 39831655 DOI: 10.1080/10408398.2025.2453632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Since the widespread usage of plastic materials and inadequate handling of plastic debris, nanoplastics (NPs) and microplastics (MPs) have become global hazards. Recent studies prove that NPs/MPs can induce various toxicities in organisms, with these adverse effects closely related to gut microbiota changes. This review thoroughly summarized the interactions between NPs/MPs and gut microbiota in various hosts, speculated on the potential factors affecting these interactions, and outlined the impacts on hosts' health caused by NPs/MPs exposure and gut microbiota dysbiosis. Firstly, different characteristics and conditions of NPs/MPs often led to complicated hazardous effects on gut microbiota. Alterations of gut microbiota composition at the phylum level were complex, while changes at the genus level exhibited a pattern of increased pathogens and decreased probiotics. Generally, the smaller size, the rougher surface, the longer shape, the higher concentration, and the longer exposure of NPs/MPs induced more severe damage to gut microbiota. Then, different adaptation and tolerance degrees of gut microbiota to NPs/MPs exposure might contribute to gut microbiota dysbiosis. Furthermore, NPs/MPs could be carriers of other hazards to generally exert more severe damage on gut microbiota. In summary, both pristine and contaminated NPs/MPs posed severe threats to hosts through inducing gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Kun Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| | - Lei Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| | - Jingyang Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| | - Xiaomeng Wu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- National Engineering Research Center for Fruit and Vegetable Processing, Key Lab of Fruit and Vegetable Processing, Ministry of Agriculture, Beijing Key Laboratory of Food Non-Thermal Processing, Beijing, China
| |
Collapse
|
32
|
Szymczak-Pajor I, Drzewoski J, Kozłowska M, Krekora J, Śliwińska A. The Gut Microbiota-Related Antihyperglycemic Effect of Metformin. Pharmaceuticals (Basel) 2025; 18:55. [PMID: 39861118 PMCID: PMC11768994 DOI: 10.3390/ph18010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
It is critical to sustain the diversity of the microbiota to maintain host homeostasis and health. Growing evidence indicates that changes in gut microbial biodiversity may be associated with the development of several pathologies, including type 2 diabetes mellitus (T2DM). Metformin is still the first-line drug for treatment of T2DM unless there are contra-indications. The drug primarily inhibits hepatic gluconeogenesis and increases the sensitivity of target cells (hepatocytes, adipocytes and myocytes) to insulin; however, increasing evidence suggests that it may also influence the gut. As T2DM patients exhibit gut dysbiosis, the intestinal microbiome has gained interest as a key target for metabolic diseases. Interestingly, changes in the gut microbiome were also observed in T2DM patients treated with metformin compared to those who were not. Therefore, the aim of this review is to present the current state of knowledge regarding the association of the gut microbiome with the antihyperglycemic effect of metformin. Numerous studies indicate that the reduction in glucose concentration observed in T2DM patients treated with metformin is due in part to changes in the biodiversity of the gut microbiota. These changes contribute to improved intestinal barrier integrity, increased production of short-chain fatty acids (SCFAs), regulation of bile acid metabolism, and enhanced glucose absorption. Therefore, in addition to the well-recognized reduction of gluconeogenesis, metformin also appears to exert its glucose-lowering effect by influencing gut microbiome biodiversity. However, we are only beginning to understand how metformin acts on specific microorganisms in the intestine, and further research is needed to understand its role in regulating glucose metabolism, including the impact of this remarkable drug on specific microorganisms in the gut.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Małgorzata Kozłowska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Jan Krekora
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| |
Collapse
|
33
|
Sternini C, Rozengurt E. Bitter taste receptors as sensors of gut luminal contents. Nat Rev Gastroenterol Hepatol 2025; 22:39-53. [PMID: 39468215 DOI: 10.1038/s41575-024-01005-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
Taste is important in the selection of food and is orchestrated by a group of distinct receptors, the taste G protein-coupled receptors (GPCRs). Taste 1 receptors (Tas1rs in mice and TAS1Rs in humans; also known as T1Rs) detect sweet and umami tastes, and taste 2 receptors (Tas2rs in mice and TAS2Rs in humans; also known as T2Rs) detect bitterness. These receptors are also expressed in extraoral sites, including the gastrointestinal mucosa. Tas2rs/TAS2Rs have gained interest as potential targets to prevent or treat metabolic disorders. These bitter taste receptors are expressed in functionally distinct types of gastrointestinal mucosal cells, including enteroendocrine cells, which, upon stimulation, increase intracellular Ca2+ and release signalling molecules that regulate gut chemosensory processes critical for digestion and absorption of nutrients, for neutralization and expulsion of harmful substances, and for metabolic regulation. Expression of Tas2rs/TAS2Rs in gut mucosa is upregulated by high-fat diets, and intraluminal bitter 'tastants' affect gastrointestinal functions and ingestive behaviour through local and gut-brain axis signalling. Tas2rs/TAS2Rs are also found in Paneth and goblet cells, which release antimicrobial peptides and glycoproteins, and in tuft cells, which trigger type 2 immune response against parasites, thus providing a direct line of defence against pathogens. This Review will focus on gut Tas2r/TAS2R distribution, signalling and regulation in enteroendocrine cells, supporting their role as chemosensors of luminal content that serve distinct functions as regulators of body homeostasis and immune response.
Collapse
Affiliation(s)
- Catia Sternini
- Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| | - Enrique Rozengurt
- Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
34
|
Ghosh S, Chorghade R, Diehl RC, Dodge GJ, Bae S, Dugan AE, Halim M, Wuo MG, Bartlett H, Herndon L, Kiessling LL, Imperiali B. Glycan analysis probes inspired by human lectins for investigating host-microbe crosstalk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.24.630132. [PMID: 39763805 PMCID: PMC11703188 DOI: 10.1101/2024.12.24.630132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Human lectins are critical carbohydrate-binding proteins that recognize diverse glycoconjugates from microorganisms and can play a key role in host-microbe interactions. Despite their importance in immune recognition and pathogen binding, the specific glycan ligands and functions of many human lectins remain poorly understood. Using previous proof-of-concept studies on selected lectins as the foundation for this work, we present ten additional glycan analysis probes (GAPs) from a diverse set of human soluble lectins, offering robust tools to investigate glycan-mediated interactions. We describe a protein engineering platform that enables scalable production of GAPs that maintain native-like conformations and oligomerization states, equipped with functional reporter tags for targeted glycan profiling. We demonstrate that the soluble GAP reagents can be used in various applications, including glycan array analysis, mucin-binding assays, tissue staining, and microbe binding in complex populations. These capabilities make GAPs valuable for dissecting interactions relevant to understanding host responses to microbes. The tools can be used to distinguish microbial from mammalian glycans, which is crucial for understanding the cross-target interactions of lectins in a physiological environment where both glycan types exist. GAPs have potential as diagnostic and prognostic tools for detecting glycan alterations in chronic diseases, microbial dysbiosis, and immune-related conditions.
Collapse
Affiliation(s)
- Soumi Ghosh
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rajeev Chorghade
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger C Diehl
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Greg J Dodge
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sunhee Bae
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amanda E Dugan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Melanie Halim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael G Wuo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Helen Bartlett
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liam Herndon
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Barbara Imperiali
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
35
|
Yu L, Qiu G, Yu X, Zhao J, Liu J, Wang H, Dong L. Terpinen-4-ol Improves the Intestinal Barrier Function of the Colon in Immune-Stressed Weaning Piglets. Animals (Basel) 2024; 15:9. [PMID: 39794952 PMCID: PMC11719020 DOI: 10.3390/ani15010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
The aim of this study was to investigate the effects of terpinen-4-ol (TER) supplementation on the intestinal barrier function of pigs. Five groups of fifty 28-day-old piglets with comparable body weights were randomly assigned to the following groups: the control group (CON), the lipopolysaccharide group (LPS), the low TER group (PLT), the middle TER group (PMT), and the high TER group (PHT). The basal diet was given to the CON and LPS groups, and 30, 60, or 90 mg/kg TER was added to the basal diet for the TER groups. After the 21-day trial period, piglets in the LPS and TER groups received an intraperitoneal injection of 100 μg/kg body weight of LPS, whereas the piglets in the CON group received an injection of 0.9% normal saline solution. The results showed that LPS stimulation resulted in a decrease (p < 0.05) in the depth of colonic crypts in piglets, which was greater (p < 0.05) in the TER group. Compared with those in the CON group, the number of goblet cells and MUC2 expression were decreased in the colon of piglets in the LPS group, while these parameters were increased in the PMT group (p < 0.05). The malondialdehyde (MDA) content was greater in the colon of the LPS group than in that of the CON group, while the activities of glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and catalase (CAT) were lower in the colon of the LPS group; conversely, the MDA content was lower in the colons of the PLT and PMT groups than in those of the LPS group (p < 0.05). TER also reduced (p < 0.05) LPS-induced upregulation of IL-1β and TNF-α expression, along with the relative gene expression of NLRP3, ASC, and caspase-1 in the colon of piglets (p < 0.05). Compared with those in the CON group, the abundances of Firmicutes and UCG-005 in the LPS group were lower (p < 0.05), and those in the TER group were significantly greater than those in the LPS group. Compared with those in the CON group, the abundance of Proteobacteria in the LPS group increased (p < 0.05), while the abundance of Actinobacteria and Phascolarctobacterium increased (p < 0.05) in the colon of the PHT group compared with that in the LPS group. In conclusion, TER effectively improved the intestinal barrier function of the colon in weaning piglets. Based on the results of this study, the appropriate dose of TER in the diets of weaning piglets was 60 mg/kg.
Collapse
Affiliation(s)
- Lihuai Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Guangzhi Qiu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Xiaomu Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Jianwei Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Jun Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Hongrong Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
| | - Li Dong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 215009, China; (L.Y.); (G.Q.); (X.Y.); (J.Z.); (J.L.); (H.W.)
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
36
|
Fortunato-Silva J, de Rezende LP, Ferreira-Neto ML, Bispo-da-Silva LB, Balbi APC. Intrauterine exposure to a high-fat diet, with different levels of lipids, and its gastrointestinal repercussions: a model of fetal programming in rats. J Dev Orig Health Dis 2024; 15:e33. [PMID: 39711030 DOI: 10.1017/s2040174424000382] [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: 12/24/2024]
Abstract
It is known that adverse stimuli, such as altered diets during pregnancy and lactation, can result in deleterious effects on the progeny. The aim of this study was to evaluate the possible gastrointestinal repercussions in the offspring of Wistar rats exposed to high-fat diets. Pregnant rats were divided into three groups: normolipidic diet (3.5% lipids), a diet containing 28% lipids, and a diet with 40% lipids. Body weight and food, water, daily caloric, and macronutrient intake were evaluated in the pregnant rats. Structural and functional gastrointestinal parameters were assessed in 30-day-old male pups. Depending on the lipid content of the maternal diet, the pups may exhibit gastric mucosal thickening, an increase in the relative weight of the small intestine, a reduction in the jejunal and ileal mucosa, and a decrease in the total thickness of the ileum. Additionally, there may be a reduction in the number of villi per area in these organs and a thinning of the muscular layer in the large intestine. The structural changes induced by the maternal high-fat diet seem to reduce the stomach's sensitivity to ethanol-induced ulcers, which is the only functional alteration observed. Therefore, the offspring of dams exposed to high-fat diets during pregnancy and lactation exhibits impaired gastrointestinal development, with alterations depending on dietary fat content and specific gastrointestinal regions. Structural changes did not always result in functional abnormalities and, in some cases, appeared protective. The long-term consequences of the observed morphological alterations require further investigation.
Collapse
Affiliation(s)
- Jéssica Fortunato-Silva
- Program in Applied Structural and Cellular Biology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Lívia Prometti de Rezende
- Program in Applied Structural and Cellular Biology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Marcos Luiz Ferreira-Neto
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Luiz Borges Bispo-da-Silva
- Department of Pharmacology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Ana Paula Coelho Balbi
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
37
|
Doranga S, Krogfelt KA, Cohen PS, Conway T. Nutrition of Escherichia coli within the intestinal microbiome. EcoSal Plus 2024; 12:eesp00062023. [PMID: 38417452 PMCID: PMC11636361 DOI: 10.1128/ecosalplus.esp-0006-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/03/2023] [Indexed: 03/01/2024]
Abstract
In this chapter, we update our 2004 review of "The Life of Commensal Escherichia coli in the Mammalian Intestine" (https://doi.org/10.1128/ecosalplus.8.3.1.2), with a change of title that reflects the current focus on "Nutrition of E. coli within the Intestinal Microbiome." The earlier part of the previous two decades saw incremental improvements in understanding the carbon and energy sources that E. coli and Salmonella use to support intestinal colonization. Along with these investigations of electron donors came a better understanding of the electron acceptors that support the respiration of these facultative anaerobes in the gastrointestinal tract. Hundreds of recent papers add to what was known about the nutrition of commensal and pathogenic enteric bacteria. The fact that each biotype or pathotype grows on a different subset of the available nutrients suggested a mechanism for succession of commensal colonizers and invasion by enteric pathogens. Competition for nutrients in the intestine has also come to be recognized as one basis for colonization resistance, in which colonized strain(s) prevent colonization by a challenger. In the past decade, detailed investigations of fiber- and mucin-degrading anaerobes added greatly to our understanding of how complex polysaccharides support the hundreds of intestinal microbiome species. It is now clear that facultative anaerobes, which usually cannot degrade complex polysaccharides, live in symbiosis with the anaerobic degraders. This concept led to the "restaurant hypothesis," which emphasizes that facultative bacteria, such as E. coli, colonize the intestine as members of mixed biofilms and obtain the sugars they need for growth locally through cross-feeding from polysaccharide-degrading anaerobes. Each restaurant represents an intestinal niche. Competition for those niches determines whether or not invaders are able to overcome colonization resistance and become established. Topics centered on the nutritional basis of intestinal colonization and gastrointestinal health are explored here in detail.
Collapse
Affiliation(s)
- Sudhir Doranga
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Karen A. Krogfelt
- Department of Science and Environment, Pandemix Center Roskilde University, Roskilde, Denmark
| | - Paul S. Cohen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, Rhode Island, USA
| | - Tyrrell Conway
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
38
|
Xu L, Li X, Han S, Mu C, Zhu W. Galacto-oligosaccharides regulate intestinal mucosal sialylation to counteract antibiotic-induced mucin dysbiosis. Food Funct 2024; 15:12016-12032. [PMID: 39563647 DOI: 10.1039/d4fo04626a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Intestinal mucin offers a physical barrier to maintain host-commensal homeostasis. Glycosylation is essential for the appropriate functioning of mucin. Galacto-oligosaccharides (GOS) have been used as a prebiotic with proven intestinal benefits, while their regulatory mechanism on mucin remains unclear. This study employed an antibiotic-treated rat model to mimic gut dysbiosis and attempted to restore gut dysbiosis using GOS. The gut microbiome and intestinal mucus O-glycosylations (O-glycans) in the small intestine were profiled by high-throughput sequencing and glycomics. The sialic acid phenotype at the end of O-glycans was further validated with lectin staining. Expressions of key enzymes in sialic acid metabolism and epithelial morphology were determined as well. Antibiotics significantly increased the relative abundance of Escherichia/Shigella and decreased the relative abundance of Lactobacillus. This was accompanied by decreased microbial sialidase activity and increased sialic acid in the digesta, as well as an increase in epithelial sialidase activity. Analysis of key sialylation enzymes showed the upregulation of α 2,6 sialylation (e.g. ST6GALNACs) and downregulation of α 2,3 sialylation (e.g. ST3GALs) after antibiotic treatment. The glycomics results revealed that antibiotics increased core 4 and α 2,6 sialylated O-glycans and decreased core 1, core 3 and α 2,3 sialylated O-glycans in the intestinal mucus of rats, which was further confirmed by lectin staining. Intestinal histology results demonstrated that antibiotic treatment led to the dysbiosis of intestinal mucus homeostasis. To further test the role of microbiota in regulating intestinal mucus sialylation, we supplemented GOS with antibiotics. The results showed that GOS reversed the effects of antibiotics on the gut microbiota and intestinal mucus O-glycans (especially sialylated O-glycans), characterized by an increase of Lactobacillus and α 2,3 sialylated O-glycans and a decrease of Escherichia/Shigella and α 2,6 sialylated O-glycans. What's more, GOS reduced the stimulation of the intestinal mucosa by lipopolysaccharide (LPS) by increasing α 2,3 sialylated intestinal alkaline phosphatase (IAP) to enhance IAP activity, thereby restoring intestinal mucus homeostasis. Overall, GOS counteracts antibiotic-induced mucin deficiency by remedying the gut ecology and changing the mucin sialylation pattern, as reflected by the increase of α 2,3 sialylated O-glycans and the decrease of α 2,6 sialylated O-glycans.
Collapse
Affiliation(s)
- Laipeng Xu
- 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
| | - Shuibing Han
- 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
| | - Chunlong Mu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB., Canada.
| | - 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
| |
Collapse
|
39
|
Trovão LDO, Vieira MAM, Santos ACDM, Puño-Sarmiento JJ, Nunes PHS, Santos FF, Rocha VGP, Knöbl T, Navarro-Garcia F, Gomes TAT. Identification of a genomic cluster related to hypersecretion of intestinal mucus and mucinolytic activity of atypical enteropathogenic Escherichia coli (aEPEC). Front Cell Infect Microbiol 2024; 14:1393369. [PMID: 39703371 PMCID: PMC11656320 DOI: 10.3389/fcimb.2024.1393369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/18/2024] [Indexed: 12/21/2024] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) strains are subdivided into typical (tEPEC) and atypical (aEPEC) according to the presence or absence of a virulence-associated plasmid called pEAF. Our research group has previously demonstrated that two aEPEC strains, 0421-1 and 3991-1, induce an increase in mucus production in a rabbit ileal loop model in vivo. This phenomenon was not observed with a tEPEC prototype strain. Few studies on aEPEC strains evaluating their capacity to induce intestinal mucus hypersecretion were done. This study aimed to investigate aEPEC strains regarding their genotypic and phenotypic characteristics, their ability to alter mucus production in an in vivo intestinal infection model, and their potential mucinolytic activity. To investigate the relationship between strains 0421-1 and 3991-1 and 11 other aEPEC strains, their serotypes, sequence types (ST), and virulence factors (VF), several sequencing and genomic analyses were carried out. The study also involved researching the reproduction of mucus hypersecretion in rabbits in vivo. We found that the two mucus-inducing strains and two other strains (1582-4 and 2531-13) shared the same phylogroup (A), ST (378), serotype (O101/O162:H33), and intimin subtype (ι2), were phylogenetically related, and induced mucus hypersecretion in vivo. A wide diversity of VFs was found among the strains, confirming their genomic heterogeneity. However, among the genes studied, no unique virulence factor or gene set was identified exclusively in the mucus-inducing strains, suggesting the multifactorial nature of this phenomenon. The two strains (1582-4 and 2531-13) closely related to the two aEPEC strains that induced mucus production in vivo also induced the phenomenon. The investigation of the mucinolytic activity revealed that all aEPEC strains used mucins as their carbon sources. Ten of the 13 aEPEC strains could cross a mucin layer, and only four adhered better to agar containing mucin than to agar without mucin. The present study paves the way for subsequent investigations into the molecular mechanisms regarding cellular interactions and responses, as well as the correlation between virulence factors and the induction of mucus production/expression during aEPEC infections.
Collapse
Affiliation(s)
- Liana de Oliveira Trovão
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mônica Aparecida Midolli Vieira
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Carolina de Mello Santos
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juan Josue Puño-Sarmiento
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Pedro Henrique Soares Nunes
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda Fernandes Santos
- Laboratório Alerta, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Terezinha Knöbl
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Fernando Navarro-Garcia
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Mexico City, Mexico
| | - Tânia Aparecida Tardelli Gomes
- Laboratório Experimental de Patogenicidade de Enterobactérias, Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
40
|
Shi H, Choppa VS, Paneru D, Kim WK. Effects of phytase and 25-Hydroxycholecalciferol supplementation in broilers fed calcium-phosphorous deficient diets, with or without Eimeria challenge, on growth performance, body composition, bone development, and gut health. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:411-428. [PMID: 39640548 PMCID: PMC11617698 DOI: 10.1016/j.aninu.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 12/07/2024]
Abstract
The study evaluated the effects of nutritional strategies on broilers challenged with Eimeria from d 14 to 26. A total of 840 Cobb male broilers were fed five diets in a 2 × 5 factorial arrangement: 1) nutrient adequate diet (PC; 0.84% calcium [Ca], 0.42% available phosphorus [avP]); 2) Ca-P deficient diet (NC; 0.64% Ca, 0.22% avP); 3) NC + 1500 FTU/kg phytase of diet (NC + PHY); 4) NC + 5000 IU/kg 25-Hydroxycholecalciferol of diet (NC + 25OHD); and 5) NC with both supplements (NC + PHY + 25OHD), with and without Eimeria challenge. All treatments had six replicate cages with 14 birds per cage. At 5 days post inoculation (DPI), the challenged birds exhibited higher serum fluorescein isothiocyanate-d (FITC-d) levels than the unchallenged birds (P < 0.001). The NC + PHY and NC + PHY + 25OHD groups exhibited lower FITC-d levels compared to the NC + 25OHD group (P = 0.012). Significant interaction effects between Eimeria challenge and dietary treatments were observed on various parameters. During 0 to 6 and 0 to 12 DPI, Eimeria challenge resulted in decreased the body weight gain (BWG) (P < 0.05) but had a negative effect on the feed conversion ratio (FCR) in birds compared to the unchallenged group (P < 0.05). Reducing Ca and avP levels in the diet (NC) did not adversely affect BWG, but negatively impacted FCR, bone ash weight, ash concentration, and femur bone microstructure parameters (P < 0.05). On 12 DPI, Eimeria challenge led to decreased tibia bone weight, bone volume, fat-free bone weight (FFBW), and ash weight of birds (P < 0.05). Supplementation with phytase alone or in combination with 25OHD improved growth performance, gut permeability, bone ash and bone microstructure parameters in birds (P < 0.05). However, the group fed 25OHD alone showed enhancements on growth performance, mineral apposition rate (MAR), bone ash concentration and ash percentage of the birds (P < 0.05). In conclusion, lowering Ca and avP levels in the diet negatively affected FCR and bone development but did not affect intestinal integrity in broilers. Dietary supplementation of phytase, 25OHD, or phytase in combination of 25OHD could enhance the growth performance and bone quality of broilers infected with Eimeria. Notably, the benefits of phytase supplementation were generally more pronounced than those associated with 25OHD supplementation; however, the combination of phytase and 25OHD could induce optimum effects.
Collapse
Affiliation(s)
- Hanyi Shi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | | | - Deependra Paneru
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Woo K. Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
41
|
Dai Z, Wang Z, Pan X, Zheng L, Xu Y, Qiao Q. Effects of triclosan adsorption on intestinal toxicity and resistance gene expression in Xenopus tropicalis with different particle sizes of polystyrene. J Environ Sci (China) 2024; 146:176-185. [PMID: 38969446 DOI: 10.1016/j.jes.2023.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2024]
Abstract
Microplastics (MPs) are commonly found with hydrophobic contaminants in the water column and pose a serious threat to aquatic organisms. The effects of polystyrene microplastics of different particle sizes on the accumulation of triclosan in the gut of Xenopus tropicalis, its toxic effects, and the transmission of resistance genes were evaluated. The results showed that co-exposure to polystyrene (PS-MPs) adsorbed with triclosan (TCS) caused the accumulation of triclosan in the intestine with the following accumulation capacity: TCS + 5 µm PS group > TCS group > TCS + 20 µm PS group > TCS + 0.1 µm PS group. All experimental groups showed increased intestinal inflammation and antioxidant enzyme activity after 28 days of exposure to PS-MPs and TCS of different particle sizes. The TCS + 20 µm PS group exhibited the highest upregulated expression of pro-inflammatory factors (IL-10, IL-1β). The TCS + 20 µm group showed the highest increase in enzyme activity compared to the control group. PS-MPs and TCS, either alone or together, altered the composition of the intestinal microbial community. In addition, the presence of more antibiotic resistance genes than triclosan resistance genes significantly increased the expression of tetracycline resistance and sulfonamide resistance genes, which may be associated with the development of intestinal inflammation and oxidative stress. This study refines the aquatic ecotoxicity assessment of TCS adsorbed by MPs and provides informative information for the management and control of microplastics and non-antibiotic bacterial inhibitors.
Collapse
Affiliation(s)
- Zhuo Dai
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zikai Wang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinying Pan
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Li Zheng
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yanbin Xu
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Qingxia Qiao
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
42
|
Guha TK, Esplin ED, Horning AM, Chiu R, Paul K, Weimer AK, Becker WR, Laquindanum R, Mills MA, Glen Esplin D, Shen J, Monte E, White S, Karathanos TV, Cotter D, Bi J, Ladabaum U, Longacre TA, Curtis C, Greenleaf WJ, Ford JM, Snyder MP. Single-cell spatial mapping reveals alteration of cell type composition and tissue microenvironment during early colorectal cancer formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.622725. [PMID: 39605357 PMCID: PMC11601668 DOI: 10.1101/2024.11.20.622725] [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: 11/29/2024]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer mortality in the United States. Familial adenomatous polyposis (FAP) is a hereditary syndrome that raises the risk of developing CRC, with total colectomy as the only effective prevention. Even though FAP is rare (0.5% of all CRC cases), this disease model is well suited for studying the early stages of malignant transformation as patients form many polyps reflective of pre-cancer states. In order to spatially profile and analyze the pre-cancer and tumor microenvironment, we have performed single-cell multiplexed imaging for 52 samples: 12 normal mucosa,16 FAP mucosa,18 FAP polyps, 2 FAP adenocarcinoma, and 4 sporadic colorectal cancer (CRCs) using Co-detection by Indexing (CODEX) imaging platform. The data revealed significant changes in cell type composition occurring in early stage polyps and during the malignant transformation of polyps to CRC. We observe a decrease in CD4+/CD8+ T cell ratio and M1/M2 macrophage ratio along the FAP disease continuum. Advanced dysplastic polyps show a higher population of cancer associated fibroblasts (CAFs), which likely alter the pre-cancer microenvironment. Within polyps and CRCs, we observe strong nuclear expression of beta-catenin and higher number neo-angiogenesis events, unlike FAP mucosa and normal colon counterparts. We identify an increase in cancer stem cells (CSCs) within the glandular crypts of the FAP polyps and also detect Tregs, tumor associated macrophages (TAMs) and vascular endothelial cells supporting CSC survival and proliferation. We detect a potential immunosuppressive microenvironment within the tumor 'nest' of FAP adenocarcinoma samples, where tumor cells tend to segregate and remain distant from the invading immune cells. TAMs were found to infiltrate the tumor area, along with angiogenesis and tumor proliferation. CAFs were found to be enriched near the inflammatory region within polyps and CRCs and may have several roles in supporting tumor growth. Neighborhood analyses between adjacent FAP mucosa and FAP polyps show significant differences in spatial location of cells based on functionality. For example, in FAP mucosa, naive CD4+ T cells alone tend to localize near the fibroblast within the stromal compartment. However, in FAP polyp, CD4+T cells colocalize with the macrophages for T cell activation. Our data are expected to serve as a useful resource for understanding the early stages of neogenesis and the pre-cancer microenvironment, which may benefit early detection, therapeutic intervention and future prevention.
Collapse
Affiliation(s)
- Tuhin K Guha
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | | | | | - Kristina Paul
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Annika K Weimer
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Meredith A Mills
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305
| | - D Glen Esplin
- Animal Reference Pathology, Salt Lake City, UT 84107
| | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA 94305
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Shannon White
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | | | - Daniel Cotter
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Joanna Bi
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| | - Uri Ladabaum
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305
| | - Teri A Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA 94305
| | - Christina Curtis
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305
| | - William J Greenleaf
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - James M Ford
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305
| |
Collapse
|
43
|
Maita K, Fujihara H, Matsumura M, Miyakawa M, Baba R, Morimoto H, Nakayama R, Ito Y, Kawaguchi K, Hamada Y. Impact of Reduced Saliva Production on Intestinal Integrity and Microbiome Alterations: A Sialoadenectomy Mouse Model Study. Int J Mol Sci 2024; 25:12455. [PMID: 39596522 PMCID: PMC11594800 DOI: 10.3390/ijms252212455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the effect of reduced saliva production on intestinal histological structure and microbiome composition using a sialoadenectomy murine model, evaluating differences in saliva secretion, body weight, intestinal histopathological changes, and microbiome alteration using 16S rRNA gene sequencing across three groups (control, sham, and sialoadenectomy). For statistical analysis, one-way analysis of variance and multiple comparisons using Bonferroni correction were performed. p-values < 0.05 were considered statistically significant. Microbiome analysis was performed using Qiime software. The results show that reduced saliva secretion leads to structural changes in the intestinal tract, including shorter and atrophic villi, deformed Paneth cells, decreased goblet cell density, and immunohistochemical changes in epidermal growth factor and poly(ADP-ribose) polymerase-1, especially at three months after surgery. They also showed significant alterations in the intestinal microbiome, including increased Lactobacillaceae and altered populations of Ruminococcaceae and Peptostreptococcaceae, suggesting potential inflammatory responses and decreased short-chain fatty acid production. However, by 12 months after surgery, these effects appeared to be normalized, indicating potential compensatory mechanisms. Interestingly, sham-operated mice displayed favorable profiles, possibly due to immune activation from minor surgical intervention. This study underscores saliva's essential role in intestinal condition, emphasizing the "oral-gut axis" and highlighting broader implications for the relationship between oral and systemic health.
Collapse
Affiliation(s)
- Kanna Maita
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
| | - Hisako Fujihara
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
- Department of Oral Hygiene, Tsurumi Junior College, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan
| | - Mitsuki Matsumura
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
| | - Moeko Miyakawa
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
| | - Ryoko Baba
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu 807-8555, Japan
| | - Hiroyuki Morimoto
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu 807-8555, Japan
| | - Ryoko Nakayama
- Department of Pathology, School of Dental Medicine, Tsurumi University 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan
| | - Yumi Ito
- Department of Diagnostic Pathology, Tsurumi University Dental Hospital, Yokohama 230-8501, Japan
| | - Koji Kawaguchi
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
| | - Yoshiki Hamada
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama 230-8501, Japan; (K.M.); (Y.H.)
| |
Collapse
|
44
|
Lv H, Park J, Lim HK, Abraham IJ, Yin X, Gao Y, Hur J. Impacts of polyhydroxybutyrate (PHB) microplastic exposure on physiology and metabolic profiles of Litopenaeus vannamei. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175588. [PMID: 39154993 DOI: 10.1016/j.scitotenv.2024.175588] [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: 05/28/2024] [Revised: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
In light of increasing concerns about microplastic pollution, it is crucial to understand the biological impacts of biodegradable PHB microplastics on marine organisms. This study included a 96-h exposure experiment to assess acute toxicity at PHB concentrations of 0 mg/L, 100 mg/L, 500 mg/L and 1000 mg/L. Additionally, a 60-day feeding trial was conducted with PHB concentrations of 0, 0.5, 1.0 and 2.0 g/kg to evaluate the long-term effects on growth, physiological health and metabolic responses of Litopenaeus vannamei. Results from the exposure experiment indicated that PHB microplastics up to 100 mg/L were non-toxic to shrimp. However, the 60-day feeding trial revealed that higher concentrations led to slight reductions in survival rates and growth performance, indicating a concentration-dependent response. Analysis of antioxidant and immune enzymes showed minimal changes across most parameters. However, increases in malondialdehyde content and lysozyme activity at higher PHB levels suggested a stress response. Microbial analysis indicated higher species richness and greater community diversity in the PHB group compared to controls, as evidenced by Chao1, ACE, Shannon and Simpson indices. Linear discriminant analysis revealed that Enterobacteriales and related taxa were more prevalent in the PHB group, while Rhodobacteraceae and associated taxa dominated the control group. Pathway analysis highlighted enhanced signal transduction, cell mobility and metabolic resource reallocation in response to PHB-induced stress. Integrated transcriptomic and metabolomic analyses revealed significant regulatory changes, especially in lipid metabolism pathways. These findings suggest that while PHB microplastics trigger adaptive metabolic responses in shrimp, they do not cause acute toxicity. Significant variations in intestinal microbiome composition reflect potential shifts in gut health dynamics due to PHB ingestion. This study enhances our understanding of the ecological impacts of microplastics and underscores the necessity for further research into the environmental safety of biodegradable alternatives.
Collapse
Affiliation(s)
- Huirong Lv
- Department of Aquaculture and Aquatic Science, Kunsan National University, Gunsan 54150, Republic of Korea
| | - Jungyeol Park
- Department of Aquaculture and Aquatic Science, Kunsan National University, Gunsan 54150, Republic of Korea
| | - Han Kyu Lim
- Interdisciplinary Program of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muan 58554, Republic of Korea
| | | | - Xiaolong Yin
- Zhoushan Fisheries Research Institute, Zhoushan, China
| | - Yang Gao
- School of Fishery, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Junwook Hur
- Department of Aquaculture and Aquatic Science, Kunsan National University, Gunsan 54150, Republic of Korea.
| |
Collapse
|
45
|
Kim M, Shin M, Zhao Y, Ghosh M, Son Y. Transformative Impact of Nanocarrier‐Mediated Drug Delivery: Overcoming Biological Barriers and Expanding Therapeutic Horizons. SMALL SCIENCE 2024; 4. [DOI: 10.1002/smsc.202400280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Advancing therapeutic progress is centered on developing drug delivery systems (DDS) that control therapeutic molecule release, ensuring precise targeting and optimal concentrations. Targeted DDS enhances treatment efficacy and minimizes off‐target effects, but struggles with drug degradation. Over the last three decades, nanopharmaceuticals have evolved from laboratory concepts into clinical products, highlighting the profound impact of nanotechnology in medicine. Despite advancements, the effective delivery of therapeutics remains challenging because of biological barriers. Nanocarriers offer a solution with a small size, high surface‐to‐volume ratios, and customizable properties. These systems address physiological and biological challenges, such as shear stress, protein adsorption, and quick clearance. They allow targeted delivery to specific tissues, improve treatment outcomes, and reduce adverse effects. Nanocarriers exhibit controlled release, decreased degradation, and enhanced efficacy. Their size facilitates cell membrane penetration and intracellular delivery. Surface modifications increase affinity for specific cell types, allowing precise treatment delivery. This study also elucidates the potential integration of artificial intelligence with nanoscience to innovate future nanocarrier systems.
Collapse
Affiliation(s)
- Minhye Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Myeongyeon Shin
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
| | - Young‐Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
- Department of Animal Biotechnology Faculty of Biotechnology College of Applied Life Sciences Jeju National University Jeju‐si Jeju Special Self‐Governing Province 63243 Republic of Korea
- Bio‐Health Materials Core‐Facility Center Jeju National University Jeju‐si 63243 Republic of Korea
- Practical Translational Research Center Jeju National University Jeju‐si 63243 Republic of Korea
| |
Collapse
|
46
|
Nwako JG, McCauley HA. Enteroendocrine cells regulate intestinal homeostasis and epithelial function. Mol Cell Endocrinol 2024; 593:112339. [PMID: 39111616 PMCID: PMC11401774 DOI: 10.1016/j.mce.2024.112339] [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: 02/29/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/11/2024]
Abstract
Enteroendocrine cells (EECs) are well-known for their systemic hormonal effects, especially in the regulation of appetite and glycemia. Much less is known about how the products made by EECs regulate their local environment within the intestine. Here, we focus on paracrine interactions between EECs and other intestinal cells as they regulate three essential aspects of intestinal homeostasis and physiology: 1) intestinal stem cell function and proliferation; 2) nutrient absorption; and 3) mucosal barrier function. We also discuss the ability of EECs to express multiple hormones, describe in vitro and in vivo models to study EECs, and consider how EECs are altered in GI disease.
Collapse
Affiliation(s)
- Jennifer G Nwako
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA
| | - Heather A McCauley
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA.
| |
Collapse
|
47
|
Jiang Q, Li Z, Dang D, Wei J, Wu H. Role of mechanosensitive channel Piezo1 protein in intestinal inflammation regulation: A potential target. FASEB J 2024; 38:e70122. [PMID: 39425504 PMCID: PMC11580726 DOI: 10.1096/fj.202401323r] [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: 06/11/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The intestine is a hollow tract that primarily transports and digests food. It often encounters mechanical forces and exotic threats, resulting in increased intestinal inflammation attributed to the consistent threat of foreign pathogens. Piezo1, a mechanosensitive ion channel, is distributed broadly and abundantly in the intestinal tissue. It transduces mechanical signals into electrochemical signals and participates in many critical life activities, such as proliferation, differentiation, cell apoptosis, immune cell activation, and migration. Its effect on inflammation has been discussed in detail in systems, such as musculoskeletal (osteoarthritis) and cardiac (myocarditis), but the effects on intestinal inflammation remain unelucidated. Piezo1 regulates mucosal layer and epithelial barrier homeostasis during the complex intestinal handling of foreign antigens and tissue trauma. It initiates and spreads immune responses and causes distant effects of inflammation in the vascular and lymphatic systems, but reports of the effects of Piezo1 in intestinal inflammation are scarce. Therefore, this study aimed to discuss the role of Piezo1 in intestinal inflammation and explore novel therapeutic targets.
Collapse
Affiliation(s)
- Qinlei Jiang
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Zhenyu Li
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Dan Dang
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Jiaqi Wei
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Hui Wu
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| |
Collapse
|
48
|
Ladaika CA, Chakraborty A, Masood A, Hostetter G, Yi JM, O'Hagan HM. LSD1 inhibition attenuates targeted therapy-induced lineage plasticity in BRAF V600E colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620306. [PMID: 39554172 PMCID: PMC11565724 DOI: 10.1101/2024.10.25.620306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
BRAF activating mutations occur in approximately 10% of metastatic colorectal cancer (CRCs) and are associated with worse prognosis due to an inferior response to standard chemotherapy. Standard of care for patients with refractory metastatic BRAF V600E CRC is treatment with BRAF and EGFR inhibitors. However, responses are not durable. Lineage plasticity to neuroendocrine cancer is an emerging mechanism of targeted therapy resistance in several cancer types. Enteroendocrine cells (EECs), the neuroendocrine cell of the intestine, are uniquely present in BRAF V600E CRC as compared to BRAF wildtype CRC. Here, we demonstrated that combined BRAF and EGFR inhibition enriches for EECs in several models of BRAF V600E CRC. Additionally, EECs and other secretory cell types were enriched in a subset of BRAF V600E CRC patient samples following targeted therapy. Importantly, inhibition of the lysine demethylase LSD1 with a clinically relevant inhibitor attenuated targeted therapy-induced EEC enrichment through blocking the interaction of LSD1, CoREST2 and STAT3. Statement of Significance Our findings that BRAF plus EGFR inhibition induces lineage plasticity in BRAF V600E CRC represents a new paradigm for how resistance to BRAF plus EGFR inhibition occurs and our finding that LSD1 inhibition blocks lineage plasticity has the potential to improve responses to BRAF plus EGFR inhibitor therapy in patients.
Collapse
|
49
|
Rezapour M, Narayanan A, Gurcan MN. Machine Learning Analysis of RNA-Seq Data Identifies Key Gene Signatures and Pathways in Mpox Virus-Induced Gastrointestinal Complications Using Colon Organoid Models. Int J Mol Sci 2024; 25:11142. [PMID: 39456924 PMCID: PMC11508207 DOI: 10.3390/ijms252011142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Mpox, caused by the Mpox virus (MPXV), emerged globally in 2022 with the Clade IIb strain, presenting a critical public health challenge. While MPXV is primarily characterized by fever and rash, gastrointestinal (GI) complications, such as diarrhea and proctitis, have also been observed. This study is a reanalysis of GSE219036 without own data and focuses on the impact of MPXV infection on the colon, using human-induced pluripotent stem cell-derived colon organoids as a model. We applied a tailored statistical framework for RNA-seq data, Generalized Linear Models with Quasi-Likelihood F-tests and Relaxed Magnitude-Altitude Scoring (GLMQL-RMAS), to identify differentially expressed genes (DEGs) across MPXV clades: MPXV I (Zr-599 Congo Basin), MPXV IIa (Liberia), and MPXV IIb (2022 MPXV). Through a novel methodology called Cross-RMAS, we ranked genes by integrating statistical significance and biological relevance across all clades. Machine learning analysis using the genes identified by Cross-RMAS, demonstrated 100% accuracy in differentiating between the different MPXV strains and mock samples. Furthermore, our findings reveal that MPXV Clade I induces the most extensive alterations in gene expression, with significant upregulation of stress response genes, such as HSPA6 and FOS, and downregulation of genes involved in cytoskeletal organization and vesicular trafficking, such as PSAP and CFL1. In contrast, Clade IIb shows the least impact on gene expression. Through Gene Ontology (GO) analysis, we identified pathways involved in protein folding, immune response, and epithelial integrity that are disrupted in infected cells, suggesting mechanisms by which MPXV may contribute to GI symptoms.
Collapse
Affiliation(s)
- Mostafa Rezapour
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| | - Aarthi Narayanan
- Department of Biology, George Mason University, Fairfax, VA 22030, USA;
| | - Metin Nafi Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| |
Collapse
|
50
|
Klingbeil EA, Schade R, Lee SH, Kirkland R, de La Serre CB. Manipulation of feeding patterns in high fat diet fed rats improves microbiota composition dynamics, inflammation and gut-brain signaling. Physiol Behav 2024; 285:114643. [PMID: 39059597 DOI: 10.1016/j.physbeh.2024.114643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Chronic consumption of high fat (HF) diets has been shown to increase meal size and meal frequency in rodents, resulting in overeating. Reducing meal frequency and establishing periods of fasting, independently of caloric intake, may improve obesity-associated metabolic disorders. Additionally, diet-driven changes in microbiota composition have been shown to play a critical role in the development and maintenance of metabolic disorders. In this study, we used a pair-feeding paradigm to reduce meal frequency and snacking episodes while maintaining overall intake and body weight in HF fed rats. We hypothesized that manipulation of feeding patterns would improve microbiota composition and metabolic outcomes. Male Wistar rats were placed in three groups consuming either a HF, low fat diet (LF, matched for sugar), or pair-fed HF diet for 7 weeks (n = 11-12/group). Pair-fed animals received the same amount of food consumed by the HF fed group once daily before dark onset (HF-PF). Rats underwent oral glucose tolerance and gut peptide cholecystokinin sensitivity tests. Bacterial DNA was extracted from the feces collected during both dark and light cycles and sequenced via Illumina MiSeq sequencing of the 16S V4 region. Our pair-feeding paradigm reduced meal numbers, especially small meals in the inactive phase, without changing total caloric intake. This shift in feeding patterns reduced relative abundances of obesity-associated bacteria and maintained circadian fluctuations in microbial abundances. These changes were associated with improved gastrointestinal (GI) function, reduced inflammation, and improved glucose tolerance and gut to brain signaling. We concluded from these data that targeting snacking may help improve metabolic outcomes, independently of energy content of the diet and hyperphagia.
Collapse
Affiliation(s)
- E A Klingbeil
- Department of Nutritional Sciences, The University of Texas at Austin, United States
| | - R Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, United States
| | - S H Lee
- Department of Food Sciences, Sun Moon University, South Korea
| | - R Kirkland
- Office of Research, University of Georgia, United States
| | - C B de La Serre
- Department of Nutritional Sciences, University of Georgia, United States; Department of Biomedical Sciences, Colorado State University, United States.
| |
Collapse
|