1
|
Dumitru A, Tocia C, Dumitru E, Stanigut AM, Cozaru GC, Matei E, Petcu LC, Popescu RC, Leopa N, Rugina S. Intestinal permeability assessed by serum zonulin in liver cirrhosis: A systematic review and meta-analysis. Medicine (Baltimore) 2025; 104:e42197. [PMID: 40258754 PMCID: PMC12014077 DOI: 10.1097/md.0000000000042197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND In recent years, increased intestinal permeability has been considered a hallmark of liver cirrhosis (LC), thereby exposing the liver to many bacteria and microbial components of a leaky gut, such as zonulin which is a recognized biomarker of intestinal permeability. The topic is underresearched and professional consensus regarding intestinal permeability in cirrhosis is still lacking. Our systematic review and meta-analysis aimed to investigate the intestinal permeability assessed by serum zonulin levels in patients with LC. METHODS The systematic search covered 3 databases with the following search key: "zonulin" AND "LC" OR "cirrhosis." Our investigated population (P) consisted of patients diagnosed with LC. Eligible studies compared the levels of serum zonulin in patients with LC (I) to the control group (C). Our primary outcome (O) was the difference in serum zonulin in patients with LC compared with controls (PROSPERO CRD42024580574). RESULTS In total, 4 studies fulfilled the eligibility criteria for the qualitative and quantitative analysis. The mean effect size estimate was 0.590 (95% confidence interval: 0.325-0.855) and statistically significant (t = 4.378, P < .001). For heterogeneity, Q-statistics and I2 values were examined. The Q-statistics (Q = 123.4974, df = 3, P < .0001) was found to be statistically significant. In addition, the I2 value was found to be 97.57% (95% confidence interval: 95.83-98.59). As a result, there was a statistically significant heterogeneity between studies. CONCLUSION In this systematic review and meta-analysis, we summarized the current evidence on the importance of zonulin. Currently, there is no clear evidence regarding the axis between the gut and liver, particularly cirrhosis, and zonulin is known as a surrogate biomarker for a leaky gut. An important aspect to consider is the fact that zonulin is primarily produced in the liver; in cases of LC, its synthesis is impaired, which is why zonulin cannot be considered a marker of intestinal permeability in this pathology. Further randomized controlled trials assessing this impaired intestinal permeability and gut-liver axis in patients with liver diseases will be necessary.
Collapse
Affiliation(s)
- Andrei Dumitru
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Cristina Tocia
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Eugen Dumitru
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
- Center for Research and development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, Constanta, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Alina Mihaela Stanigut
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Georgeta Camelia Cozaru
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
- Center for Research and development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, Constanta, Romania
| | - Elena Matei
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
- Center for Research and development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, Constanta, Romania
| | | | - Razvan Catalin Popescu
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Nicoleta Leopa
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- County Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Sorin Rugina
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
- Clinical Hospital of Infectious Diseases Constanta, Constanta, Romania
| |
Collapse
|
2
|
Linsalata M, Prospero L, Ignazzi A, Riezzo G, D’Attoma B, Mallardi D, Goscilo F, Notarnicola M, De Nunzio V, Pinto G, Russo F. Depression in Diarrhea-Predominant IBS Patients: Exploring the Link Between Gut Barrier Dysfunction and Erythrocyte Polyunsaturated Fatty Acid Levels. J Clin Med 2025; 14:2483. [PMID: 40217932 PMCID: PMC11989550 DOI: 10.3390/jcm14072483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Patients with irritable bowel syndrome (IBS) often experience comorbid psychological conditions, notably depression and anxiety. Evidence suggests that these conditions are linked to gut barrier dysfunction, dysbiosis, and chronic inflammation. All these factors are central to IBS pathophysiology and mood disturbances. Polyunsaturated fatty acids (PUFAs) play crucial roles in modulating inflammation and depression. This study examined the associations among intestinal permeability, PUFA profiles, low-grade inflammation, and depression severity in IBS patients with diarrhea (IBS-D). Methods: Forty-three IBS-D patients (7 men, 36 women; 44.56 ± 1.52 years) were categorized into depressed (IBS-D(d+)) and non-depressed (IBS-D(d-)) groups according to scores on the depression subscale of the Symptom Checklist-90-Revised (SCL-90-R). Biomarkers of small intestinal permeability (s-IP) were assessed in urine and blood, alongside erythrocyte membrane PUFA composition, dysbiosis, and inflammation indices. Results: IBS-D (d+) patients exhibited elevated s-IP and altered PUFA metabolism compared to their IBS-D (d-) counterparts. Additionally, in the first group, omega-3 PUFA concentrations inversely correlated with s-IP biomarkers, while the omega-6/omega-3 ratio showed a positive correlation. Moreover, depression severity is significantly associated with s-IP markers and omega-3 PUFA levels. Lastly, IBS-D (d+) patients exhibited higher levels of dysbiosis and pro-inflammatory cytokines than IBS-D (d-) patients. Conclusions: These findings highlight the interplay between intestinal barrier integrity and PUFA metabolism in IBS-D patients with depression, suggesting that s-IP markers and erythrocyte PUFA profiles could represent novel therapeutic targets for managing depression in this population. This study was registered on ClinicalTrials.gov (NCT03423069), with a date of registration of 30 January 2018.
Collapse
Affiliation(s)
- Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Laura Prospero
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Antonia Ignazzi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Giuseppe Riezzo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Francesco Goscilo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Valentina De Nunzio
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Giuliano Pinto
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (L.P.); (A.I.); (G.R.); (B.D.); (D.M.); (F.G.)
| |
Collapse
|
3
|
Westmark CJ. Soy-based purified ingredient diet affects mouse gut permeability and the microbiome in fragile X mice. Front Mol Neurosci 2025; 18:1520211. [PMID: 40190341 PMCID: PMC11968763 DOI: 10.3389/fnmol.2025.1520211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Gastrointestinal problems including vomiting, reflux, flatulence, diarrhea, constipation and colic are common comorbidities in fragile X syndrome. There is accumulating evidence suggesting that leaky gut syndrome causes neurological phenotypes. Although fragile X messenger ribonucleoprotein is ubiquitously expressed, there is a dearth of knowledge regarding its role outside of the brain including effects on gut dysfunction in fragile X. The aim of this study was to generate novel data on gastrointestinal barrier function and the gut microbiome in response to Fmr1 genotype, sex and diet in mice. Methods Fmr1KO male mice and littermate controls in an FVB background were maintained on two purified ingredient diets (AIN-93G with casein protein versus soy protein isolate) versus two standard chows (Teklad 2019 with wheat, corn and yeast protein versus Purina 5015 with wheat, soy, corn, yeast and whey protein sources). Gut permeability was quantified by FITC-dextran levels in blood plasma. The cecal microbiome was identified by 16S rRNA sequencing. In addition, gut permeability was tested in Fmr1KO mice in the C57BL/6 J background maintained on casein- and soy protein isolate-based AIN-93G versus Teklad 2019. Results Knockout of the Fmr1 gene in FVB mice did not affect gut permeability. Soy protein isolate-based AIN-93G increased gut permeability. Beta-diversity of the cecal microbiome was significantly altered as a function of the four test diets. Akkermansia_muciniphila was increased in Fmr1KO mice fed AIN-93G while unnamed species within the genus Anaerovorax and family Ruminococcaceae were increased and the order Clostridales decreased in Fmr1KO mice fed AIN-93G/soy. Fmr1KO mice in the C57BL/6 J background exhibited increased gut permeability in response to soy protein. Discussion These findings regarding the effects of diet on gut permeability and the microbiome have important implications for experimental design. Single-source diets are ubiquitously used to maintain laboratory animals for medical research and feed details are frequently not reported in publications. Diet/phenotype interactions could have a large impact on inter-laboratory replicability in premedical research. For infants with fragile X, early-life diet could impact the severity of disease outcomes.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
4
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 DOI: 10.1186/s13054-025-05309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
5
|
Ramal-Sanchez M, Bravo-Trippetta C, D’Antonio V, Corvaglia E, Kämpfer AAM, Schins RPF, Serafini M, Angelino D. Development and assessment of an intestinal tri-cellular model to investigate the pro/anti-inflammatory potential of digested foods. Front Immunol 2025; 16:1545261. [PMID: 39975553 PMCID: PMC11835836 DOI: 10.3389/fimmu.2025.1545261] [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: 12/14/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Introduction Immunonutrition, defined as the potential of foods, nutrients and dietary patterns to modulate the immune system activity, has been proposed as a strategy to enhance the immune response in both metabolic and immune-mediated diseases. However, the anti-/pro-inflammatory role of foods and diets is far to be fully ascertained, and thus there is a continued needed for appropriate in vitro cell-culture models to investigate the role of foods in modulating cell-mediated inflammatory processes. This study aims to develop and test an in vitro tri-culture model, simulating the complexity of the intestinal tract and its multiple cell interactions. Methods To achieve this, the intestinal epithelial barrier was established by co-culturing human Caco-2 enterocyte-like and HT29-MTX-E12 mucus producing goblet-like colon cells, then adding human monocyte THP-1 cells to the basolateral compartment. The integrity and stability of the epithelial barrier were monitored and the inflammatory response of the model was assessed using various stressors at different concentrations, both individually and in combination (phorbol-12- myristate-13-acetate or PMA, and lipopolysaccharide or LPS), in terms of cytokines production. To test the model, different concentrations of in vitro digested broccoli (BD) were added to the apical section of the model. Results Supernatants from the basolateral compartment were collected and analyzed for cytokines production (IL-6, TNF-α, IL-12p70, IL-18 and IL-8) using automated ELISA (ELLA). Additionally, ZO-1 protein from the tight junctions of epithelial cells was analyzed by flow cytometry. The results indicated that 100 nM PMA added to the whole model for 20 h was the best stressor to simulate a mild-inflammatory status of the gut. Following treatment with BD, IL-6, TNF-α, IL-8 and IL-18 were significantly reduced compared to the control group, while ZO-1 expression increased at the lowest BD concentration. Conclusions These findings confirm the feasibility of the model for assessing the effects of food digesta on specific cytokines and permeability markers, representing a valuable strategy for investigating the role of foods in modulating the inflammatory response. The results obtained may support dietary strategies aimed at promoting wellbeing and preventing inflammatory-related metabolic diseases.
Collapse
Affiliation(s)
- Marina Ramal-Sanchez
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Chiara Bravo-Trippetta
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Veronica D’Antonio
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Elena Corvaglia
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Angela A. M. Kämpfer
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Roel P. F. Schins
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Mauro Serafini
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Donato Angelino
- Functional Foods and Stress Prevention Laboratory, Department of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
6
|
van Beek N, Katavisto I, Lehto M, Kolho KL, de Vos WM, Salonen A, Korpela K. Host-microbiota interactions in the infant gut revealed by daily faecal sample time series. MICROBIOME RESEARCH REPORTS 2024; 4:13. [PMID: 40207273 PMCID: PMC11977378 DOI: 10.20517/mrr.2024.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 04/11/2025]
Abstract
Aim: This study aims to explore the interplay between host immune factors and gut microbiota in human infants in vivo using time-series daily stool samples and identify biomarkers of host-microbe interactions. Methods: 216 faecal samples collected from infants aged 5-6 or 11-12 months were analysed for gut microbiota composition, total bacterial load, and biomarkers of immune function. Results: We identified indications of microbial stimulation of eosinophil cationic protein (ECP), IgA, calprotectin (Cal), intestinal alkaline phosphatase (IAP), and Bactericidal/permeability-increasing protein (BPI) at 6 and 12 months, as well as stimulation of lipocalin 2 (LCN2), lactoferrin (LTF), and alpha-defensin-5 only at 6 months. The associations between biomarker concentrations and bacterial population growth were primarily positive at 6 months and mostly negative at 12 months, suggesting increasing host regulation of the microbiota with age. The exceptions were IAP, which was predictive of declining bacterial populations at both time points, and Cal, whose associations changed from negative at 6 months to positive at 12 months. Conclusion: There is an age-associated development in the correlation pattern between bacterial population growth and the biomarker concentrations, suggesting that host-microbe interactions change during early development. Albumin appeared as a potential marker of gut permeability, while LCN2 seemed to correlate with gut transit time. Mucin degradation appeared to decrease with age. Mucin2 and IAP emerged as potentially important regulators of the bacterial populations in the infant gut. The study demonstrates the utility of biomarker and bacteria profiling from daily stool samples for analysing in vivo associations between the immune system and the gut microbiota and provides evidence of host regulation of the microbiota in infants.
Collapse
Affiliation(s)
- Nienke van Beek
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Iiris Katavisto
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki 00250, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki 00014, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Kaija-Leena Kolho
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Faculty of Medicine, University of Helsinki and Children’s Hospital, Helsinki University Hospital HUS, Helsinki 00014, Finland
| | - Willem M. de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen 6700 EH, the Netherlands
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
- Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsink 00014, Finland
| |
Collapse
|
7
|
Li K, Wei W, Xu C, Lian X, Bao J, Yang S, Wang S, Zhang X, Zheng X, Wang Y, Zhong S. Prebiotic inulin alleviates anxiety and depression-like behavior in alcohol withdrawal mice by modulating the gut microbiota and 5-HT metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156181. [PMID: 39488100 DOI: 10.1016/j.phymed.2024.156181] [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: 06/30/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Alcohol dependence (AD) is a common psychiatric disorder, often accompanied by anxiety and depression. These comorbidities are linked to disturbances in serotonin (5-HT) metabolism and gut microbiota dysbiosis. Clinical studies suggest that inulin, a prebiotic, can alleviate anxiety and depression in AD patients by affecting the gut microbiota, although the mechanisms remain unclear. PURPOSE The purpose of this study is to investigate the potential mechanisms by which inulin, a prebiotic, improves anxiety and depression-like behaviors in AD withdrawal mice. This research is based on the drug and food homology and intestinal treatment of encephalopathy, with the goal of developing new clinical strategies for AD treatment. STUDY DESIGN For this purpose, fecal samples from AD patients were analyzed to identify microorganisms associated with AD. An AD withdrawal mouse model was created, with inulin as the intervention and fluvoxamine maleate as the control. Techniques such as 16S microbiome sequencing and UPLC-TQMS-targeted metabolomics were used to assess gut microbiota, short-chain fatty acids (SCFAs) levels, and 5-HT metabolism. METHODS The AD withdrawal model was built using the "Drinking-in-the-dark" protocol over 6 weeks. Inulin (2 g/kg/day) and fluvoxamine maleate (30 mg/kg/day) were administered for 4 weeks. The open field test, forced swim test, and tail suspension test were used to evaluate anxiety and depression-like behaviors in mice. ELISA and qRT-PCR assessed 5-HT metabolism in the colon, blood, and prefrontal cortex, while 16S microbiome sequencing analyzed changes in gut microbiota and UPLC-TQMS examined SCFAs levels. Immunohistochemistry was used to study intestinal barrier integrity. RESULTS AD patients showed reduced SCFA-producing bacteria such as Faecalibacterium and Roseburia. In mice, AD withdrawal led to anxiety and depression-like behaviors, disrupted 5-HT metabolism, and gut microbiota dysbiosis. Inulin supplementation alleviated these behaviors, increased 5-HT and 5-hydroxytryptophan (5-HTP) levels, upregulated colonic tryptophan hydroxylase 1 (TPH1) expression, and promoted the growth of beneficial bacteria such as Faecalibacterium and Roseburia, while also increasing SCFAs levels. CONCLUSION Inulin increases the abundance of Faecalibacterium and Roseburia, enhances SCFAs production, and regulates 5-HT metabolism, improving anxiety and depression-like behaviors in AD withdrawal mice. These findings suggest that inulin may serve as a nutritional intervention for mental health in AD patients by targeting the microbiome-gut-brain axis.
Collapse
Affiliation(s)
- Kuan Li
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Wei Wei
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Chongchong Xu
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Xinqing Lian
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Jianjun Bao
- Department of Geriatric Psychiatry, The Mental Hospital of Yunnan Province, Kunming, 650224, China
| | - Shuo Yang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Shixu Wang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Xulan Zhang
- Department of Psychiatry/Alcohol Dependence Treatment, The Mental Hospital of Yunnan Province, Kunming, 650224, China
| | - Xinjian Zheng
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Yue Wang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Shurong Zhong
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China; NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China; Forensic Biology Identification Laboratory, Judicial Identification Center of Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
8
|
Borkent J, Ioannou M, Neijzen D, Haarman BCM, Sommer IEC. Probiotic Formulation for Patients With Bipolar or Schizophrenia Spectrum Disorder: A Double-Blind, Randomized Placebo-Controlled Trial. Schizophr Bull 2024:sbae188. [PMID: 39504580 DOI: 10.1093/schbul/sbae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
BACKGROUND AND HYPOTHESIS Probiotic augmentation offers a promising treatment for bipolar disorder (BD) and schizophrenia spectrum disorder (SSD). By targeting microbiome deviations, they may improve both gut and brain health. STUDY DESIGN In this double-blind, randomized, placebo-controlled trial with the multi-strain probiotic formulation Ecologic BARRIER, we aimed to improve psychiatric and cognitive symptoms, intestinal permeability, and gastrointestinal symptoms in patients with BD or SSD. A total of 131 patients were randomized 1:1 to receive either the probiotic supplement (n = 67) or a placebo (n = 64) for 3 months, in addition to treatment-as-usual. The primary outcomes were symptom severity assessed by the Brief Psychiatric Rating Scale and cognitive functioning by the Brief Assessment of Cognition in Schizophrenia. STUDY RESULTS No significant effect of probiotics was observed on psychiatric symptoms, but borderline significant improvement was observed in the cognition category of verbal memory (Linear Mixed Model (LMM) 0.33; adjusted P = .059). Probiotics beneficially affected markers of intestinal permeability and inflammation, including zonulin (LMMserum = -18.40; adjusted P = .002; LMMfecal = -10.47; adjusted P = .014) and alpha-1 antitrypsin (LMM 9.26; adjusted P = .025). Indigestion complaints significantly decreased in male participants in the probiotics group (LMM = -0.70; adjusted P = .010). Adverse events were similar between groups. CONCLUSIONS Our study observed significant advantages of probiotics for gut health in BD and SSD, with excellent safety and tolerability. A borderline effect on verbal memory was also indicated. These results underscore the need for further research into microbiome-targeted interventions for patients with complex brain disorders.
Collapse
Affiliation(s)
- Jenny Borkent
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Magdalini Ioannou
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Dorien Neijzen
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| | - Iris E C Sommer
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
9
|
Ahmad Fadzuli NI, Lim SM, Neoh CF, Majeed ABA, Tan MP, Khor HM, Tan AH, Ramasamy K. Faecal intestinal permeability and intestinal inflammatory markers in older adults with age-related disorders: A systematic review and meta-analysis. Ageing Res Rev 2024; 101:102506. [PMID: 39306247 DOI: 10.1016/j.arr.2024.102506] [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: 05/28/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
This systematic review and meta-analysis appraised previous findings to uncover potential faecal intestinal permeability and intestinal inflammatory markers in older adults. A comprehensive literature search led to the identification of ten eligible studies with findings of potential faecal intestinal permeability (zonulin and alpha-1-antitrypsin) and intestinal inflammatory markers [calprotectin, lactoferrin and neutrophil gelatinase-associated lipocalin (NGAL)]. Most of the cases (n > 2) [Parkinson's disease (PD) and Alzheimer's disease (AD)] exhibited higher faecal alpha-1-antitrypsin, zonulin and calprotectin levels. The present meta-analysis confirmed significantly higher faecal alpha-1-antitrypsin in older persons with PD compared to non-PD [MD = 22.92 mg/dL; 95 % CI = 14.02-31.81, p < 0.00001; I2 = 0 % (p = 0.73)]. There was, however, no significant difference in faecal zonulin between PD and non-PD individuals [MD = 26.88 ng/mL; 95 % CI = -29.26-83.01, p = 0.35; I2 = 94 % (p < 0.0001)]. Meanwhile, faecal calprotectin was higher in older adults with GI symptoms, multiple system atrophy (MSA) or PD than the healthy controls [MD = 9.51 μg/g; 95 % CI = 0.07-18.95, p = 0.05; I2 = 84 % (p < 0.00001)]. Altogether, faecal calprotectin appears to be a potential intestinal inflammatory marker whereas previous findings on faecal alpha-1-antitrypsin as an intestinal permeability marker remain limited and require further validation.
Collapse
Affiliation(s)
- Nurul Izzati Ahmad Fadzuli
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor Darul Ehsan 42300, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor Darul Ehsan 42300, Malaysia
| | - Chin Fen Neoh
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor Darul Ehsan 42300, Malaysia
| | - Abu Bakar Abdul Majeed
- Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, University Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, Selangor Darul Ehsan, Selangor Darul Ehsan 42300, Malaysia
| | - Maw Pin Tan
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Hui Min Khor
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Ai Huey Tan
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, Bandar Puncak Alam, Selangor Darul Ehsan 42300, Malaysia.
| |
Collapse
|
10
|
Al-Akayleh F, Agha ASAA, Al-Remawi M, Al-Adham ISI, Daadoue S, Alsisan A, Khattab D, Malath D, Salameh H, Al-Betar M, AlSakka M, Collier PJ. What We Know About the Actual Role of Traditional Probiotics in Health and Disease. Probiotics Antimicrob Proteins 2024; 16:1836-1856. [PMID: 38700762 DOI: 10.1007/s12602-024-10275-7] [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] [Accepted: 04/22/2024] [Indexed: 10/02/2024]
Abstract
The complex relationship between probiotics and human health goes beyond their traditional function in gut health, generating considerable interest for their broad potential in disease treatment. This review explores the various functions of probiotics, highlighting their impact on the immune system, their benefits for gut and oral health, their effects on metabolic and neurological disorders, and their emerging potential in cancer therapy. We give significant importance to studying the effects of probiotics on the gut-brain axis, revealing new and non-invasive therapeutic approaches for complex neurological disorders. In addition, we expand the discussion to encompass the impact of probiotics on the gut-liver and gut-lung axes, recognizing their systemic effects and potential in treating respiratory and hepatic conditions. The use of probiotic "cocktails" to improve cancer immunotherapy outcomes indicates a revolutionary approach to oncological treatments. The review explores the specific benefits associated with various strains and the genetic mechanisms that underlie them. This study sets the stage for precision medicine, where probiotic treatments can be tailored to meet the unique needs of each patient. Recent developments in delivery technologies, including microencapsulation and nanotechnology, hold great potential for enhancing the effectiveness and accuracy of probiotic applications in therapeutic settings. This study provides a strong basis for future scientific research and clinical use, promoting the incorporation of probiotics into treatment plans for a wide range of diseases. This expands our understanding of the potential benefits of probiotics in modern medicine.
Collapse
Affiliation(s)
- Faisal Al-Akayleh
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan.
| | - Ahmed S A Ali Agha
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
- Faculty of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Mayyas Al-Remawi
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Ibrahim S I Al-Adham
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Saifeddin Daadoue
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Anagheem Alsisan
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Dana Khattab
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Doha Malath
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Haneen Salameh
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Maya Al-Betar
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Motaz AlSakka
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Phillip J Collier
- Faculty of Pharmacy & Medical Sciences, University of Petra, Amman, 11196, Jordan.
| |
Collapse
|
11
|
Górecka A, Jura-Półtorak A, Koźma EM, Szeremeta A, Olczyk K, Komosińska-Vassev K. Biochemical Modulators of Tight Junctions (TJs): Occludin, Claudin-2 and Zonulin as Biomarkers of Intestinal Barrier Leakage in the Diagnosis and Assessment of Inflammatory Bowel Disease Progression. Molecules 2024; 29:4577. [PMID: 39407507 PMCID: PMC11478261 DOI: 10.3390/molecules29194577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Considering the increasing worldwide prevalence of inflammatory bowel disease (IBD), the early diagnosis of this disease is extremely important. However, non-invasive diagnostic methods remain limited, while invasive techniques are the most commonly used in daily practice. Therefore, there is a serious need to find new non-invasive biomarkers of IBD. METHODS The serum profiles of occludin, claudin-2, and zonulin were assessed in IBD patients using the ELISA method. The levels of the analyzed biomarkers were measured before and after a year of anti-inflammatory treatment, which was a tumor necrosis factor α (TNF-α) inhibitor (adalimumab) in patients with ulcerative colitis (UC) and conventional therapy in patients with Crohn's disease (CD). RESULTS In IBD patients, the serum level of occludin (p < 0.001) decreased compared to healthy individuals, while the level of claudin-2 (p < 0.001) increased. Additionally, zonulin (p < 0.01) concentration increased in CD patients compared to the control group. The highest diagnostic ability was presented by occludin measurements with the area under the curve (AUC) of 0.959 (95% CI 0.907-1) in UC and 0.948 (95% CI 0.879-1) in CD. Claudin-2 also demonstrated very good ability in diagnosing UC and CD with AUC values of 0.864 (95% CI 0.776-0.952) and 0.896 (95% CI 0.792-0.999), respectively. The ability of zonulin to diagnose CD was estimated as good with an AUC of 0.74 (95% CI 0.598-0.881). Moreover, a significant correlation was identified between C-reactive protein (CRP), claudin-2 (r = -0.37; p < 0.05), and zonulin (r = -0.44; p < 0.05) in UC patients. Treatment with adalimumab improved the level of occludin, claudin-2, and zonulin in UC patients, while anti-inflammatory conventional therapy decreased the concentration of zonulin in CD. CONCLUSIONS Occludin and claudin-2 measurements present significant utility in diagnosing both UC and CD, while zonulin assessments may be useful in CD diagnosis. Additionally, claudin-2 and zonulin measurements may be helpful in evaluating the intensity of the inflammatory process. Anti-TNF-α treatment improved the value of occludin, claudin-2, and zonulin, indicating its beneficial effect on the integrity of tight junctions in UC.
Collapse
Affiliation(s)
- Aleksandra Górecka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.J.-P.); (E.M.K.); (A.S.); (K.O.); (K.K.-V.)
| | | | | | | | | | | |
Collapse
|
12
|
Cui Y, Zhang M, Wang H, Yu T, Zhang A, Lin G, Guo Y, Wu Y. Organic Trace Minerals Enhance the Gut Health of British Shorthair Cats by Regulating the Structure of Intestinal Microbiota. Metabolites 2024; 14:494. [PMID: 39330501 PMCID: PMC11434296 DOI: 10.3390/metabo14090494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Trace minerals are essential for biological processes, including enzyme function, immune response, and hormone synthesis. The study assessed the effects of different dietary trace minerals on the gut health, microbiota composition, and immune function of cats. Eighteen adult British Shorthair cats were divided into three groups receiving inorganic trace minerals (ITM), a 50/50 mix of inorganic and organic trace minerals (ITM + OTM), or organic trace minerals (OTM) for 28 days. The OTM showed enhanced immune capacities, reduced intestinal barrier function, and lower inflammation condition. The OTM altered gut microbiota diversity, with a lower Simpson index and higher Shannon index (p < 0.05). Specifically, the abundance of Bacteroidota, Lachnospiraceae, and Prevotella in the OTM group were higher than the ITM group (p < 0.05). Metabolomic analysis identified 504 differential metabolites between the OTM and ITM groups (p < 0.05, VIP-pred-OPLS-DA > 1), affecting pathways related to steroid hormone biosynthesis and glycerophospholipid metabolism (p < 0.05, VIP-pred-OPLS-DA > 2). Additionally, there was a significant correlation between intestinal microbiota and differential metabolites. To conclude, dietary OTM can modulate the gut metabolite and microbiota composition, enhance immune and intestinal barrier function, and mitigate inflammation in cats, highlighting the benefit of using OTM in feline diet to promote the intestinal and overall health.
Collapse
Affiliation(s)
- Yingyue Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Tong Yu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Anxuan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| | - Gang Lin
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (G.L.); (Y.G.)
| | - Yuhan Guo
- Beijing Alltech Biological Products (China) Co., Ltd., Beijing 100600, China; (G.L.); (Y.G.)
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.C.); (M.Z.); (H.W.); (T.Y.); (A.Z.)
| |
Collapse
|
13
|
Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol 2024; 15:1434804. [PMID: 39301033 PMCID: PMC11410641 DOI: 10.3389/fimmu.2024.1434804] [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: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
This comprehensive review undertakes a multidisciplinary exploration of the gut-lung axis, from the foundational aspects of anatomy, embryology, and histology, through the functional dynamics of pathophysiology, to implications for clinical science. The gut-lung axis, a bidirectional communication pathway, is central to understanding the interconnectedness of the gastrointestinal- and respiratory systems, both of which share embryological origins and engage in a continuous immunological crosstalk to maintain homeostasis and defend against external noxa. An essential component of this axis is the mucosa-associated lymphoid tissue system (MALT), which orchestrates immune responses across these distant sites. The review delves into the role of the gut microbiome in modulating these interactions, highlighting how microbial dysbiosis and increased gut permeability ("leaky gut") can precipitate systemic inflammation and exacerbate respiratory conditions. Moreover, we thoroughly present the implication of the axis in oncological practice, particularly in lung cancer development and response to cancer immunotherapies. Our work seeks not only to synthesize current knowledge across the spectrum of science related to the gut-lung axis but also to inspire future interdisciplinary research that bridges gaps between basic science and clinical application. Our ultimate goal was to underscore the importance of a holistic understanding of the gut-lung axis, advocating for an integrated approach to unravel its complexities in human health and disease.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Viktória Halasy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csenge Somodi
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Mógor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
14
|
Guido D, Maqoud F, Aloisio M, Mallardi D, Ura B, Gualandi N, Cocca M, Russo F. Transcriptomic Module Discovery of Diarrhea-Predominant Irritable Bowel Syndrome: A Causal Network Inference Approach. Int J Mol Sci 2024; 25:9322. [PMID: 39273274 PMCID: PMC11394741 DOI: 10.3390/ijms25179322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Irritable bowel syndrome with diarrhea (IBS-D) is the most prevalent subtype of IBS, characterized by chronic gastrointestinal symptoms in the absence of identifiable pathological findings. This study aims to investigate the molecular mechanisms underlying IBS-D using transcriptomic data. By employing causal network inference methods, we identify key transcriptomic modules associated with IBS-D. Utilizing data from public databases and applying advanced computational techniques, we uncover potential biomarkers and therapeutic targets. Our analysis reveals significant molecular alterations that affect cellular functions, offering new insights into the complex pathophysiology of IBS-D. These findings enhance our understanding of the disease and may foster the development of more effective treatments.
Collapse
Affiliation(s)
- Davide Guido
- Data Science Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Bari, Italy
| | - Fatima Maqoud
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Bari, Italy
| | - Michelangelo Aloisio
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Bari, Italy
| | - Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Bari, Italy
| | - Blendi Ura
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy
| | - Nicolò Gualandi
- Department of Medicine, Laboratory of Biochemistry, University of Udine, P.le Kolbe 4, 33100 Udine, Italy
| | - Massimiliano Cocca
- INSERM U1052, CNRS UMR_5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France
- Institute of Hepatology Lyon (IHL), 69002 Lyon, France
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Bari, Italy
| |
Collapse
|
15
|
Linsalata M, Ignazzi A, D’Attoma B, Riezzo G, Mallardi D, Orlando A, Prospero L, Notarnicola M, De Nunzio V, Pinto G, Russo F. Relationship between Markers of Gut Barrier Function and Erythrocyte Membrane PUFAs in Diarrhea-Predominant IBS Patients Undergoing a Low-FODMAP Diet. Nutrients 2024; 16:2706. [PMID: 39203842 PMCID: PMC11357486 DOI: 10.3390/nu16162706] [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: 06/24/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Many patients with irritable bowel syndrome (IBS) have a compromised intestinal barrier associated with low-grade inflammation. Polyunsaturated fatty acids (PUFAs) are potential mediators of inflammation: omega-6 PUFAs are pro-inflammatory, while omega-3 PUFAs are antioxidant and anti-inflammatory. Zonulin is a potential biomarker for small intestinal permeability (s-IP). This study investigated the relationship between PUFAs and gastrointestinal (GI) barrier integrity in IBS patients with predominant diarrhea (IBS-D). We evaluated GI barrier function indicators in the urine and bloodstream and erythrocyte membrane PUFA composition in 38 IBS-D patients (5 men, 33 women, 44.11 ± 1.64 years), categorized at baseline by fecal zonulin levels into high (≥107 ng/mL, H-FZ) and normal (<107 ng/mL N-FZ) groups. Evaluations were conducted prior to and following a 12-week diet low in FODMAPs (LFD). At baseline, H-FZ patients had s-IP significantly higher than the reference value, lower n-3 PUFAs levels, and higher n-6/n-3 PUFAs and arachidonic acid (AA) to eicosapentaenoic acid (EPA) ratios than N-FZ. After LFD, H-FZ patients showed significant increases in n-3 PUFAs levels; decreases in n-6 PUFAs, n-6/n-3 PUFAs and AA/EPA ratios; and improved s-IP. The n-6/n-3 PUFAs ratio positively correlated with fecal zonulin levels in all subjects. These findings highlight the relationship between PUFAs and the intestinal barrier, suggesting their role in IBS-D pathophysiology and confirming the positive effects of LFD in managing IBS-D.
Collapse
Affiliation(s)
- Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Antonia Ignazzi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Giuseppe Riezzo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Antonella Orlando
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Laura Prospero
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Valentina De Nunzio
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Giuliano Pinto
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.N.); (V.D.N.); (G.P.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “S. de Bellis”, 70013 Castellana Grotte, Italy; (M.L.); (A.I.); (B.D.); (G.R.); (D.M.); (A.O.); (L.P.)
| |
Collapse
|
16
|
Mickiewicz-Góra D, Sznurkowska K, Skonieczna-Żydecka K, Drozd A, Borkowska A, Zagierski M, Troch J, Szlagatys-Sidorkiewicz A. Markers of Intestinal Permeability and Inflammation in Enterally Fed Children with Cerebral Palsy. Nutrients 2024; 16:2447. [PMID: 39125328 PMCID: PMC11314602 DOI: 10.3390/nu16152447] [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: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Cerebral palsy (CP) results in non-progressive damage to the central nervous system, leading to functional disorders of the gastrointestinal tract and requiring enteral nutrition via gastrostomy in some patients. The aim of the study was to assess the impact of enteral nutrition on intestinal inflammation expressed by stool calprotectin and intestinal permeability determined by fecal zonulin and IFABP, and to determine whether CP affects these parameters. The study group consisted of 30 children with CP, fed enterally (Cerebral Palsy Enteral Nutrition-CPEN), and two reference groups: 24 children with CP, fed orally with a standard diet (CPC-Cerebral Palsy Controls) and 24 healthy children (HC-healthy controls). The differences between these groups and between the combined CP groups (CPG and CPEN + CPC) and HC were analyzed. Fecal zonulin, calprotectin, and intestinal fatty acid-binding protein 2 (IFABP2) levels were determined by ELISA. The concentrations of fecal calprotectin and zonulin were significantly higher in the CPEN group than in the CPC group (p = 0.012, p = 0.025). When comparing the CPG (n = 53) with the HC group (n = 24), statistically significant differences were observed for calprotectin (p = 0.000018, higher in the CPG) and IFABP (p = 0.021, higher in HC). Enteral nutrition was associated in our cohort with increased fecal calprotectin and zonulin. Children with cerebral palsy presented with increased fecal calprotectin but not increased intestinal permeability expressed by stool zonulin.
Collapse
Affiliation(s)
- Dorota Mickiewicz-Góra
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Katarzyna Sznurkowska
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | | | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland;
| | - Anna Borkowska
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Maciej Zagierski
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Joanna Troch
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Agnieszka Szlagatys-Sidorkiewicz
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| |
Collapse
|
17
|
Hagemeyer H, Hellwinkel OJC, Plata-Bello J. Zonulin as Gatekeeper in Gut-Brain Axis: Dysregulation in Glioblastoma. Biomedicines 2024; 12:1649. [PMID: 39200114 PMCID: PMC11352073 DOI: 10.3390/biomedicines12081649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/01/2024] Open
Abstract
Novel biomarkers and therapeutic strategies for glioblastoma, the most common malignant brain tumor with an extremely unfavorable prognosis, are urgently needed. Recent studies revealed a significant upregulation of the protein zonulin in glioblastoma, which correlates with patient survival. Originally identified as pre-haptoglobin-2, zonulin modulates both the intestinal barrier and the blood-brain barrier by disassembling tight junctions. An association of zonulin with various neuroinflammatory diseases has been observed. It can be suggested that zonulin links a putative impairment of the gut-brain barrier with glioblastoma carcinogenesis, leading to an interaction of the gut microbiome, the immune system, and glioblastoma. We therefore propose three interconnected hypotheses: (I) elevated levels of zonulin in glioblastoma contribute to its aggressiveness; (II) upregulated (serum-) zonulin increases the permeability of the microbiota-gut-brain barrier; and (III) this creates a carcinogenic and immunosuppressive microenvironment preventing the host from an effective antitumor response. The role of zonulin in glioblastoma highlights a promising field of research that could yield diagnostic and therapeutic options for glioblastoma patients and other diseases with a disturbed microbiota-gut-brain barrier.
Collapse
Affiliation(s)
- Hannah Hagemeyer
- Institut für Neuroimmunologie und Multiple Sklerose, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany;
| | - Olaf J. C. Hellwinkel
- Department of Forensic Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251 Hamburg, Germany
| | - Julio Plata-Bello
- Department of Neurosurgery, Hospital Universitario de Canarias, S/C de Tenerife, 38320 La Laguna, Spain
| |
Collapse
|
18
|
Bai F, Bono V, Borghi L, Bonazza F, Falcinella C, Vitaletti V, Miraglia F, Trunfio M, Calcagno A, Cusato J, Vegni E, d’Arminio Monforte A, Marchetti G. Association between tight junction proteins and cognitive performance in untreated persons with HIV. AIDS 2024; 38:1292-1303. [PMID: 38704619 PMCID: PMC11216391 DOI: 10.1097/qad.0000000000003923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/29/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND HIV-associated neurocognitive disorders (HAND) still affects persons with HIV (PWH) and their pathogenesis is not completely understood. We aimed to explore the association between plasma and cerebrospinal fluid (CSF) markers of blood-brain barrier (BBB) impairment and HAND in untreated PWH. DESIGN Cross-sectional study. METHODS We enrolled untreated PWH, who underwent blood examinations and lumbar puncture to measure inflammation (IL-15, TNF-α), BBB damage (zonulin and tight junction proteins, tight junction proteins: occludin, claudin-5) and endothelial adhesion molecules (VCAM-1, ICAM-1). A comprehensive neurocognitive battery was used to diagnose HAND (Frascati criteria). RESULTS Twenty-one patients (21/78, 26.9%) patients presented HAND (100% ANI). HAND patients displayed more frequently non-CNS AIDS-defining conditions, lower nadir CD4 + T cells and increased CD4 + T-cell exhaustion (lower CD4 + CD127 + and CD4 + CD45RA + T-cell percentages), in comparison to individuals without cognitive impairment. Furthermore, HAND was characterized by higher plasma inflammation (IL-15) but lower CSF levels of biomarkers of BBB impairment (zonulin and occludin). The association between BBB damage with HAND was confirmed by fitting a multivariable logistic regression. CSF/plasma endothelial adhesion molecules were not associated with HAND but with a poor performance in different cognitive domains. CONCLUSION By showing heightened inflammation and BBB impairment, our study suggests loss of BBB integrity as a possible factor contributing to the development of HAND in untreated PWH.
Collapse
Affiliation(s)
| | | | - Lidia Borghi
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Federica Bonazza
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Camilla Falcinella
- Unit of Infectious Diseases, ASST della Valle Olona, Busto Arsizio Hospital, Busto Arsizio
| | | | | | | | | | - Jessica Cusato
- Laboratory of Pharmacology and Pharmacotherapy, Amedeo di Savoia Hospital, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elena Vegni
- Unit of Clinical Psychology, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | | | | |
Collapse
|
19
|
Misiak B, Pawlak E, Rembacz K, Kotas M, Żebrowska-Różańska P, Kujawa D, Łaczmański Ł, Piotrowski P, Bielawski T, Samochowiec J, Samochowiec A, Karpiński P. Associations of gut microbiota alterations with clinical, metabolic, and immune-inflammatory characteristics of chronic schizophrenia. J Psychiatr Res 2024; 171:152-160. [PMID: 38281465 DOI: 10.1016/j.jpsychires.2024.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/31/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
The present study had the following aims: 1) to compare gut microbiota composition in patients with schizophrenia and controls and 2) to investigate the association of differentially abundant bacterial taxa with markers of inflammation, intestinal permeability, lipid metabolism, and glucose homeostasis as well as clinical manifestation. A total of 115 patients with schizophrenia during remission of positive and disorganization symptoms, and 119 controls were enrolled. Altogether, 32 peripheral blood markers were assessed. A higher abundance of Eisenbergiella, Family XIII AD3011 group, Eggerthella, Hungatella, Lactobacillus, Olsenella, Coprobacillus, Methanobrevibacter, Ligilactobacillus, Eubacterium fissicatena group, and Clostridium innocuum group in patients with schizophrenia was found. The abundance of Paraprevotella and Bacteroides was decreased in patients with schizophrenia. Differentially abundant genera were associated with altered levels of immune-inflammatory markers, zonulin, lipid profile components, and insulin resistance. Moreover, several correlations of differentially abundant genera with cognitive impairment, higher severity of negative symptoms, and worse social functioning were observed. The association of Methanobrevibacter abundance with the level of negative symptoms, cognition, and social functioning appeared to be mediated by the levels of interleukin-6 and RANTES. In turn, the association of Hungatella with the performance of attention was mediated by the levels of zonulin. The findings indicate that compositional alterations of gut microbiota observed in patients with schizophrenia correspond with clinical manifestation, intestinal permeability, subclinical inflammation, lipid profile alterations, and impaired glucose homeostasis. Subclinical inflammation and impaired gut permeability might mediate the association of gut microbiota alterations with psychopathological symptoms and cognitive impairment.
Collapse
Affiliation(s)
- Błażej Misiak
- Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland.
| | - Edyta Pawlak
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Krzysztof Rembacz
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marek Kotas
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Paulina Żebrowska-Różańska
- Laboratory of Genomics & Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dorota Kujawa
- Laboratory of Genomics & Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Łukasz Łaczmański
- Laboratory of Genomics & Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Patryk Piotrowski
- Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| | - Tomasz Bielawski
- Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University, Szczecin, Poland
| | - Agnieszka Samochowiec
- Department of Clinical Psychology, Institute of Psychology, University of Szczecin, Poland
| | - Paweł Karpiński
- Laboratory of Genomics & Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland; Department of Genetics, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
20
|
Yang W, Xi C, Yao H, Yuan Q, Zhang J, Chen Q, Wu G, Hu J. Oral administration of lysozyme protects against injury of ileum via modulating gut microbiota dysbiosis after severe traumatic brain injury. Front Cell Infect Microbiol 2024; 14:1304218. [PMID: 38352055 PMCID: PMC10861676 DOI: 10.3389/fcimb.2024.1304218] [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: 09/29/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
Objective The current study sought to clarify the role of lysozyme-regulated gut microbiota and explored the potential therapeutic effects of lysozyme on ileum injury induced by severe traumatic brain injury (sTBI) and bacterial pneumonia in vivo and in vitro experiments. Methods Male 6-8-week-old specific pathogen-free (SPF) C57BL/6 mice were randomly divided into Normal group (N), Sham group (S), sTBI group (T), sTBI + or Lysozyme-treated group (L), Normal + Lysozyme group (NL) and Sham group + Lysozyme group (SL). At the day 7 after establishment of the model, mice were anesthetized and the samples were collected. The microbiota in lungs and fresh contents of the ileocecum were analyzed. Lungs and distal ileum were used to detect the degree of injury. The number of Paneth cells and the expression level of lysozyme were assessed. The bacterial translocation was determined. Intestinal organoids culture and co-coculture system was used to test whether lysozyme remodels the intestinal barrier through the gut microbiota. Results After oral administration of lysozyme, the intestinal microbiota is rebalanced, the composition of lung microbiota is restored, and translocation of intestinal bacteria is mitigated. Lysozyme administration reinstates lysozyme expression in Paneth cells, thereby reducing intestinal permeability, pathological score, apoptosis rate, and inflammation levels. The gut microbiota, including Oscillospira, Ruminococcus, Alistipes, Butyricicoccus, and Lactobacillus, play a crucial role in regulating and improving intestinal barrier damage and modulating Paneth cells in lysozyme-treated mice. A co-culture system comprising intestinal organoids and brain-derived proteins (BP), which demonstrated that the BP effectively downregulated the expression of lysozyme in intestinal organoids. However, supplementation of lysozyme to this co-culture system failed to restore its expression in intestinal organoids. Conclusion The present study unveiled a virtuous cycle whereby oral administration of lysozyme restores Paneth cell's function, mitigates intestinal injury and bacterial translocation through the remodeling of gut microbiota.
Collapse
Affiliation(s)
- Weijian Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Caihua Xi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haijun Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jun Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Qifang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Department of Neurosurgery and Neurocritical Care, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| |
Collapse
|
21
|
Bonaz B. The gut-brain axis in Parkinson's disease. Rev Neurol (Paris) 2024; 180:65-78. [PMID: 38129277 DOI: 10.1016/j.neurol.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
There is a bi-directional communication between the gut, including the microbiota, and the brain through the autonomic nervous system. Accumulating evidence has suggested a bidirectional link between gastrointestinal inflammation and neurodegeneration, in accordance with the concept of the gut-rain axis. An abnormal microbiota-gut-brain interaction contributes to the pathogeny of Parkinson's disease. This supports the hypothesis that Parkinson's disease originates in the gut to spread to the central nervous system, in particular through the vagus nerve. Targeting the gut-to-brain axis with vagus nerve stimulation, fecal microbiota transplantation, gut-selective antibiotics, as well as drugs targeting the leaky gut might be of interest in the management of Parkinson's disease.
Collapse
Affiliation(s)
- B Bonaz
- Service d'hépato-gastroentérologie, Grenoble institut neurosciences, université Grenoble-Alpes, Grenoble, France.
| |
Collapse
|
22
|
Chen X, Hu H, Lin X, Chen M, Bao W, Wu Y, Li C, Gao Y, Hou S, Yang Q, Chen L, Zhang J, Chen K, Wang Q, Zhu A. Euphorbia factor L1 inhibited transport channel and energy metabolism in human colon adenocarcinoma cell line Caco-2. Biomed Pharmacother 2023; 169:115919. [PMID: 37992574 DOI: 10.1016/j.biopha.2023.115919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023] Open
Abstract
Euphorbia factor L1 (EFL1) is a kind of lathyrane-type diterpenoid and is isolated from the medical herb Euphorbia lathyris L. (Euphorbiaceae); it has been reported with the toxicity that causes intestinal irritation, but the underlying mechanisms are still obscure. The objective of this study was to assess the EFL1-induced intestinal cytotoxicity in human colon adenocarcinoma Caco-2 cells. The Caco-2 cells were treated with EFL1, and the intracellular calcium ion concentration, mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (mPTP), adenosine 5'-triphosphate (ATP) content, ATPase activities, TGF-β1 concentration, and transepithelial electrical resistance (TEER) were detected. The interaction between EFL1 and the tight junction proteins Occludin, Claudin-4, Tricellulin, ZO-1, JAM-1, and E-cadherin was simulated by molecular docking. The expression of proteins involved in the energy metabolism, the ion transporters and aquaporins, the tight junction, and the F-actin cytoskeleton were detected by Western blotting and cell immunofluorescence. As a result, EFL1 decreased the intracellular Ca2+, MMP, mPTP, ATP content, and ATPase activities in the Caco-2 cells. The AMPK/SIRT1/PGC-1α signaling pathway, which regulates the energy metabolism, was inhibited. The ion transporters NEH and CFTR, as well as the aquaporins in the Caco-2 cells, were decreased. The tight junction proteins were down-regulated, and the integrity of the intestinal barrier was injured; TGF-β1 was compensatively increased; so, the intestinal permeability was increased and was characterized by decreased TEER. The morphology of the F-actin cytoskeleton was destroyed. These findings indicated that EFL1 caused cytotoxicity in the human intestinal Caco-2 cells through mitochondrial damage, inhibition of the energy metabolism, and suppression of the ion and water molecule transporters, as well as the down-regulation tight junction and cytoskeleton protiens.
Collapse
Affiliation(s)
- Xiaoying Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Hong Hu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Xiaohuang Lin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Mengting Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Wenqiang Bao
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Yajiao Wu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Chutao Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Yadong Gao
- Fujian Provincial Key Laboratory of Zoonosis Research, Fujian Center for Disease Control and Prevention, Fuzhou 350001, China; Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qiaomei Yang
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Li Chen
- Department of Gynecology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, China
| | - Jian Zhang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China.
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China.
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China.
| |
Collapse
|
23
|
Zingone F, Bertin L, Maniero D, Palo M, Lorenzon G, Barberio B, Ciacci C, Savarino EV. Myths and Facts about Food Intolerance: A Narrative Review. Nutrients 2023; 15:4969. [PMID: 38068827 PMCID: PMC10708184 DOI: 10.3390/nu15234969] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Most adverse reactions to food are patient self-reported and not based on validated tests but nevertheless lead to dietary restrictions, with patients believing that these restrictions will improve their symptoms and quality of life. We aimed to clarify the myths and reality of common food intolerances, giving clinicians a guide on diagnosing and treating these cases. We performed a narrative review of the latest evidence on the widespread food intolerances reported by our patients, giving indications on the clinical presentations, possible tests, and dietary suggestions, and underlining the myths and reality. While lactose intolerance and hereditary fructose intolerance are based on well-defined mechanisms and have validated diagnostic tests, non-coeliac gluten sensitivity and fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) intolerance are mainly based on patients' reports. Others, like non-hereditary fructose, sorbitol, and histamine intolerance, still need more evidence and often cause unnecessary dietary restrictions. Finally, the main outcome of the present review is that the medical community should work to reduce the spread of unvalidated tests, the leading cause of the problematic management of our patients.
Collapse
Affiliation(s)
- Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy;
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy;
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
| | - Michela Palo
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
| | - Brigida Barberio
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy;
| | - Carolina Ciacci
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Salerno, Italy;
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35124 Padua, Italy; (L.B.); (D.M.); (M.P.); (G.L.); (E.V.S.)
- Gastroenterology Unit, Azienda Ospedale—Università Padova, 35128 Padua, Italy;
| |
Collapse
|
24
|
Guiducci L, Cabiati M, Santocchi E, Prosperi M, Morales MA, Muratori F, Randazzo E, Federico G, Calderoni S, Del Ry S. Expression of miRNAs in Pre-Schoolers with Autism Spectrum Disorders Compared with Typically Developing Peers and Its Effects after Probiotic Supplementation. J Clin Med 2023; 12:7162. [PMID: 38002774 PMCID: PMC10672692 DOI: 10.3390/jcm12227162] [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: 10/25/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Alteration of the microbiota-gut-brain axis has been recently recognized as a possible contributor to the physiopathology of autism spectrum disorder (ASD). In this context, microRNA (miRNAs) dysfunction, implicated both in several neuropathological conditions including ASD and in different gastrointestinal disorders (GIDs), could represent an important modulating factor. In this contextual framework, we studied the transcriptional profile of specific circulating miRNAs associated with both ASD (miR-197-5p, miR-424-5p, miR-500a-5p, miR-664a-5p) and GID (miR-21-5p, miR-320a-5p, miR-31-5p, miR-223-5p) in a group of pre-schoolers with ASD and in typically developing (TD) peers. In the ASD group, we also assessed the same miRNAs after a 6-month supplementation with probiotics and their correlation with plasma levels of zonulin and lactoferrin. At baseline, the expression of miRNAs involved in ASD were significantly reduced in ASD pre-schoolers vs. TD controls. Regarding the miRNAs involved in GID, the expression levels of miR-320-5p, miR-31-5p, and miR-223-5p were significantly higher in ASD than in TD subjects, whereas miR-21-5p showed significantly reduced expression in the ASD group vs. TD group. Supplementation with probiotics did not significantly change the expression of miRNAs in the ASD population. We found a significative negative correlation between zonulin and miR-197-5p and miR-21-5p at baseline, as well as between lactoferrin and miR-223-5p after 6 months of probiotic supplementation. Our study confirms the presence of an altered profile of the miRNAs investigated in ASD versus TD peers that was not modified by supplementation with probiotics.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Manuela Cabiati
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Elisa Santocchi
- UFSMIA Zona Valle del Serchio, Azienda USL Toscana Nord Ovest, 55032 Castelnuovo di Garfagnana, Italy;
| | - Margherita Prosperi
- UFSMIA Valdera-Alta Val di Cecina, Azienda USL Toscana Nord Ovest, 56128 Pisa, Italy;
| | - Maria Aurora Morales
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Filippo Muratori
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Emioli Randazzo
- Unit of Pediatric Endocrinology and Diabetes, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (E.R.); (G.F.)
| | - Giovanni Federico
- Unit of Pediatric Endocrinology and Diabetes, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (E.R.); (G.F.)
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Ry
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| |
Collapse
|
25
|
VanElzakker MB, Tillman EM, Yonker LM, Ratai EM, Georgiopoulos AM. Neuropsychiatric adverse effects from CFTR modulators deserve a serious research effort. Curr Opin Pulm Med 2023; 29:603-609. [PMID: 37655981 PMCID: PMC10552811 DOI: 10.1097/mcp.0000000000001014] [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] [Indexed: 09/02/2023]
Abstract
PURPOSE OF REVIEW This review highlights the problem of neuropsychiatric adverse effects (AEs) associated with elexacaftor/tezacaftor/ivacaftor (ETI), current suboptimal mitigation approaches, a novel testable mechanistic hypothesis, and potential solutions requiring further research. RECENT FINDINGS Studies show that a minority of persons with cystic fibrosis (PwCF) initiating cystic fibrosis transmembrane conductance regulator (CFTR) modulators experience neuropsychiatric AEs including worsening mood, cognition, anxiety, sleep, and suicidality. The GABA-A receptor is a ligand-gated chloride channel, and magnetic resonance spectroscopy neuroimaging studies have shown that reduced GABA expression in rostral anterior cingulate cortex is associated with anxiety and depression. Recent research details the impact of peripheral inflammation and the gut-brain axis on central neuroinflammation. Plasma ETI concentrations and sweat chloride have been evaluated in small studies of neuropsychiatric AEs but not validated to guide dose titration or correlated with pharmacogenomic variants or safety/efficacy. SUMMARY Although ETI is well tolerated by most PwCF, some experience debilitating neuropsychiatric AEs. In some cases, these AEs may be driven by modulation of CFTR and chloride transport within the brain. Understanding biological mechanisms is a critical next step in identifying which PwCF are likely to experience AEs, and in developing evidence-based strategies to mitigate them, while retaining modulator efficacy.
Collapse
|
26
|
Ohtani N, Kamiya T, Kawada N. Recent updates on the role of the gut-liver axis in the pathogenesis of NAFLD/NASH, HCC, and beyond. Hepatol Commun 2023; 7:e0241. [PMID: 37639702 PMCID: PMC10462074 DOI: 10.1097/hc9.0000000000000241] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/30/2023] [Indexed: 08/31/2023] Open
Abstract
The gut and the liver are anatomically and physiologically connected, and this connection is called the "gut-liver axis," which exerts various influences on liver physiology and pathology. The gut microbiota has been recognized to trigger innate immunity and modulate the liver immune microenvironment. Gut microbiota influences the physiological processes in the host, such as metabolism, by acting on various signaling receptors and transcription factors through their metabolites and related molecules. The gut microbiota has also been increasingly recognized to modulate the efficacy of immune checkpoint inhibitors. In this review, we discuss recent updates on gut microbiota-associated mechanisms in the pathogenesis of chronic liver diseases such as NAFLD and NASH, as well as liver cancer, in light of the gut-liver axis. We particularly focus on gut microbial metabolites and components that are associated with these liver diseases. We also discuss the role of gut microbiota in modulating the response to immunotherapy in liver diseases.
Collapse
Affiliation(s)
- Naoko Ohtani
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Tomonori Kamiya
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Norifumi Kawada
- Department of Hepatology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| |
Collapse
|